WO2019234221A1 - Methods for stratification and treatment of a patient suffering from chronic lymphocytic leukemia - Google Patents
Methods for stratification and treatment of a patient suffering from chronic lymphocytic leukemia Download PDFInfo
- Publication number
- WO2019234221A1 WO2019234221A1 PCT/EP2019/064946 EP2019064946W WO2019234221A1 WO 2019234221 A1 WO2019234221 A1 WO 2019234221A1 EP 2019064946 W EP2019064946 W EP 2019064946W WO 2019234221 A1 WO2019234221 A1 WO 2019234221A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- patient
- foxp3
- chronic lymphocytic
- tgfp
- Prior art date
Links
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 title claims abstract description 176
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 title claims abstract description 124
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 title claims abstract description 118
- 238000000034 method Methods 0.000 title claims abstract description 97
- 238000011282 treatment Methods 0.000 title claims abstract description 62
- 238000013517 stratification Methods 0.000 title abstract description 6
- 101000861452 Homo sapiens Forkhead box protein P3 Proteins 0.000 claims abstract description 125
- 102100027581 Forkhead box protein P3 Human genes 0.000 claims abstract description 119
- 102000003814 Interleukin-10 Human genes 0.000 claims abstract description 119
- 108090000174 Interleukin-10 Proteins 0.000 claims abstract description 119
- 230000000750 progressive effect Effects 0.000 claims abstract description 73
- 150000001875 compounds Chemical class 0.000 claims description 65
- 239000003112 inhibitor Substances 0.000 claims description 63
- 239000012472 biological sample Substances 0.000 claims description 30
- 238000011394 anticancer treatment Methods 0.000 claims description 9
- 210000004027 cell Anatomy 0.000 abstract description 288
- 230000014509 gene expression Effects 0.000 abstract description 91
- 102000004127 Cytokines Human genes 0.000 abstract description 58
- 108090000695 Cytokines Proteins 0.000 abstract description 58
- 206010028980 Neoplasm Diseases 0.000 abstract description 53
- 108090000623 proteins and genes Proteins 0.000 abstract description 52
- 210000001744 T-lymphocyte Anatomy 0.000 abstract description 46
- 230000001105 regulatory effect Effects 0.000 abstract description 31
- 102000004169 proteins and genes Human genes 0.000 abstract description 26
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 abstract description 25
- 210000003289 regulatory T cell Anatomy 0.000 abstract description 22
- 238000004519 manufacturing process Methods 0.000 abstract description 21
- 201000010099 disease Diseases 0.000 abstract description 19
- 102000037983 regulatory factors Human genes 0.000 abstract description 11
- 108091008025 regulatory factors Proteins 0.000 abstract description 11
- 230000028327 secretion Effects 0.000 abstract description 10
- 230000002596 correlated effect Effects 0.000 abstract description 8
- 239000003550 marker Substances 0.000 abstract description 6
- 210000002707 regulatory b cell Anatomy 0.000 abstract description 6
- 230000001684 chronic effect Effects 0.000 abstract description 5
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 abstract description 4
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 abstract description 4
- 230000001506 immunosuppresive effect Effects 0.000 abstract description 4
- 230000000527 lymphocytic effect Effects 0.000 abstract description 4
- 208000012526 B-cell neoplasm Diseases 0.000 abstract description 3
- 230000008685 targeting Effects 0.000 abstract description 3
- 206010062016 Immunosuppression Diseases 0.000 abstract description 2
- 230000024245 cell differentiation Effects 0.000 abstract description 2
- 230000004663 cell proliferation Effects 0.000 abstract description 2
- 238000010230 functional analysis Methods 0.000 abstract description 2
- 208000026278 immune system disease Diseases 0.000 abstract description 2
- 210000000227 basophil cell of anterior lobe of hypophysis Anatomy 0.000 abstract 1
- 239000000090 biomarker Substances 0.000 description 90
- 239000000427 antigen Substances 0.000 description 45
- 241000282414 Homo sapiens Species 0.000 description 44
- 210000003719 b-lymphocyte Anatomy 0.000 description 43
- 101000934341 Homo sapiens T-cell surface glycoprotein CD5 Proteins 0.000 description 42
- 102100025244 T-cell surface glycoprotein CD5 Human genes 0.000 description 42
- 108091007433 antigens Proteins 0.000 description 42
- 102000036639 antigens Human genes 0.000 description 42
- 239000000523 sample Substances 0.000 description 37
- 201000011510 cancer Diseases 0.000 description 35
- 239000011324 bead Substances 0.000 description 33
- 238000004458 analytical method Methods 0.000 description 29
- 239000000203 mixture Substances 0.000 description 23
- 235000018102 proteins Nutrition 0.000 description 23
- 230000000694 effects Effects 0.000 description 22
- 238000000684 flow cytometry Methods 0.000 description 21
- 239000003814 drug Substances 0.000 description 20
- -1 CP- 547632 Chemical compound 0.000 description 17
- 108091023037 Aptamer Proteins 0.000 description 16
- 150000001413 amino acids Chemical class 0.000 description 16
- 101000998953 Homo sapiens Immunoglobulin heavy variable 1-2 Proteins 0.000 description 15
- 108010009583 Transforming Growth Factors Proteins 0.000 description 15
- 102000009618 Transforming Growth Factors Human genes 0.000 description 15
- 239000002955 immunomodulating agent Substances 0.000 description 15
- 108020004999 messenger RNA Proteins 0.000 description 15
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 14
- 108010029697 CD40 Ligand Proteins 0.000 description 14
- 102100032937 CD40 ligand Human genes 0.000 description 14
- 102100036887 Immunoglobulin heavy variable 1-2 Human genes 0.000 description 14
- 230000006698 induction Effects 0.000 description 14
- 230000000638 stimulation Effects 0.000 description 14
- 102100026008 Breakpoint cluster region protein Human genes 0.000 description 13
- 108090000994 Catalytic RNA Proteins 0.000 description 13
- 102000053642 Catalytic RNA Human genes 0.000 description 13
- 210000004369 blood Anatomy 0.000 description 13
- 239000008280 blood Substances 0.000 description 13
- 239000003795 chemical substances by application Substances 0.000 description 13
- 229940079593 drug Drugs 0.000 description 13
- 239000012634 fragment Substances 0.000 description 13
- 108091092562 ribozyme Proteins 0.000 description 13
- 230000004083 survival effect Effects 0.000 description 13
- 108060003951 Immunoglobulin Proteins 0.000 description 12
- 241001465754 Metazoa Species 0.000 description 12
- 230000000875 corresponding effect Effects 0.000 description 12
- 102000018358 immunoglobulin Human genes 0.000 description 12
- 230000001965 increasing effect Effects 0.000 description 12
- 230000000770 proinflammatory effect Effects 0.000 description 12
- 239000000243 solution Substances 0.000 description 12
- 229940121358 tyrosine kinase inhibitor Drugs 0.000 description 12
- 239000013598 vector Substances 0.000 description 12
- 238000009169 immunotherapy Methods 0.000 description 11
- 239000013612 plasmid Substances 0.000 description 11
- 238000002560 therapeutic procedure Methods 0.000 description 11
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 11
- 102100031585 ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Human genes 0.000 description 10
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 10
- 101000777636 Homo sapiens ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Proteins 0.000 description 10
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 10
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 10
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 10
- 108091034117 Oligonucleotide Proteins 0.000 description 10
- 238000003501 co-culture Methods 0.000 description 10
- 238000005516 engineering process Methods 0.000 description 10
- 238000011002 quantification Methods 0.000 description 10
- 238000012360 testing method Methods 0.000 description 10
- 102100027207 CD27 antigen Human genes 0.000 description 9
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 9
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 9
- 102000014150 Interferons Human genes 0.000 description 9
- 108010050904 Interferons Proteins 0.000 description 9
- 238000011319 anticancer therapy Methods 0.000 description 9
- 238000003556 assay Methods 0.000 description 9
- 238000000338 in vitro Methods 0.000 description 9
- 238000002955 isolation Methods 0.000 description 9
- 238000012423 maintenance Methods 0.000 description 9
- 150000007523 nucleic acids Chemical class 0.000 description 9
- 150000003839 salts Chemical class 0.000 description 9
- 206010061818 Disease progression Diseases 0.000 description 8
- 102000001398 Granzyme Human genes 0.000 description 8
- 108060005986 Granzyme Proteins 0.000 description 8
- 108010004729 Phycoerythrin Proteins 0.000 description 8
- 108700012920 TNF Proteins 0.000 description 8
- 241000700605 Viruses Species 0.000 description 8
- 239000012190 activator Substances 0.000 description 8
- 239000002671 adjuvant Substances 0.000 description 8
- 238000001514 detection method Methods 0.000 description 8
- 230000005750 disease progression Effects 0.000 description 8
- 230000006870 function Effects 0.000 description 8
- 210000000987 immune system Anatomy 0.000 description 8
- 229940047124 interferons Drugs 0.000 description 8
- 239000003446 ligand Substances 0.000 description 8
- 210000001165 lymph node Anatomy 0.000 description 8
- 101710117290 Aldo-keto reductase family 1 member C4 Proteins 0.000 description 7
- 108020004414 DNA Proteins 0.000 description 7
- 241000702421 Dependoparvovirus Species 0.000 description 7
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 7
- 238000001793 Wilcoxon signed-rank test Methods 0.000 description 7
- 239000002246 antineoplastic agent Substances 0.000 description 7
- 239000000074 antisense oligonucleotide Substances 0.000 description 7
- 238000012230 antisense oligonucleotides Methods 0.000 description 7
- 230000008901 benefit Effects 0.000 description 7
- 229940127089 cytotoxic agent Drugs 0.000 description 7
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 7
- 238000007417 hierarchical cluster analysis Methods 0.000 description 7
- 230000004957 immunoregulator effect Effects 0.000 description 7
- 238000004949 mass spectrometry Methods 0.000 description 7
- 230000007246 mechanism Effects 0.000 description 7
- 238000003199 nucleic acid amplification method Methods 0.000 description 7
- 102000039446 nucleic acids Human genes 0.000 description 7
- 108020004707 nucleic acids Proteins 0.000 description 7
- 238000002360 preparation method Methods 0.000 description 7
- 108090000765 processed proteins & peptides Proteins 0.000 description 7
- 230000004044 response Effects 0.000 description 7
- 239000000126 substance Substances 0.000 description 7
- 150000004917 tyrosine kinase inhibitor derivatives Chemical class 0.000 description 7
- 241001430294 unidentified retrovirus Species 0.000 description 7
- 101150013553 CD40 gene Proteins 0.000 description 6
- 102000003964 Histone deacetylase Human genes 0.000 description 6
- 108090000353 Histone deacetylase Proteins 0.000 description 6
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 6
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 6
- 230000003321 amplification Effects 0.000 description 6
- 238000003776 cleavage reaction Methods 0.000 description 6
- 230000001419 dependent effect Effects 0.000 description 6
- 208000035475 disorder Diseases 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 238000009472 formulation Methods 0.000 description 6
- 230000012010 growth Effects 0.000 description 6
- 230000003053 immunization Effects 0.000 description 6
- 230000002401 inhibitory effect Effects 0.000 description 6
- 239000012528 membrane Substances 0.000 description 6
- 239000002245 particle Substances 0.000 description 6
- 239000013610 patient sample Substances 0.000 description 6
- 230000002285 radioactive effect Effects 0.000 description 6
- 230000003439 radiotherapeutic effect Effects 0.000 description 6
- 230000007017 scission Effects 0.000 description 6
- 239000007787 solid Substances 0.000 description 6
- 238000011285 therapeutic regimen Methods 0.000 description 6
- 238000001262 western blot Methods 0.000 description 6
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 5
- 101100112922 Candida albicans CDR3 gene Proteins 0.000 description 5
- 102000004190 Enzymes Human genes 0.000 description 5
- 108090000790 Enzymes Proteins 0.000 description 5
- 108010002350 Interleukin-2 Proteins 0.000 description 5
- 102000000588 Interleukin-2 Human genes 0.000 description 5
- 102100030704 Interleukin-21 Human genes 0.000 description 5
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 5
- 230000009471 action Effects 0.000 description 5
- 238000007792 addition Methods 0.000 description 5
- 238000003491 array Methods 0.000 description 5
- 210000001185 bone marrow Anatomy 0.000 description 5
- 238000002512 chemotherapy Methods 0.000 description 5
- 239000006185 dispersion Substances 0.000 description 5
- 239000000975 dye Substances 0.000 description 5
- AAKJLRGGTJKAMG-UHFFFAOYSA-N erlotinib Chemical compound C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 AAKJLRGGTJKAMG-UHFFFAOYSA-N 0.000 description 5
- 230000028993 immune response Effects 0.000 description 5
- 238000002649 immunization Methods 0.000 description 5
- 108010074108 interleukin-21 Proteins 0.000 description 5
- 238000007918 intramuscular administration Methods 0.000 description 5
- 238000002372 labelling Methods 0.000 description 5
- 210000005210 lymphoid organ Anatomy 0.000 description 5
- 238000002493 microarray Methods 0.000 description 5
- 210000005259 peripheral blood Anatomy 0.000 description 5
- 239000011886 peripheral blood Substances 0.000 description 5
- 239000000047 product Substances 0.000 description 5
- 208000037821 progressive disease Diseases 0.000 description 5
- 230000010076 replication Effects 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- 238000007920 subcutaneous administration Methods 0.000 description 5
- 230000001629 suppression Effects 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 4
- 102000007644 Colony-Stimulating Factors Human genes 0.000 description 4
- 108010071942 Colony-Stimulating Factors Proteins 0.000 description 4
- 238000002965 ELISA Methods 0.000 description 4
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 4
- 108090000978 Interleukin-4 Proteins 0.000 description 4
- 102000015696 Interleukins Human genes 0.000 description 4
- 108010063738 Interleukins Proteins 0.000 description 4
- 241001529936 Murinae Species 0.000 description 4
- 101100268066 Mus musculus Zap70 gene Proteins 0.000 description 4
- 239000002253 acid Substances 0.000 description 4
- 230000000890 antigenic effect Effects 0.000 description 4
- 239000007864 aqueous solution Substances 0.000 description 4
- 230000004071 biological effect Effects 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 229940047120 colony stimulating factors Drugs 0.000 description 4
- 239000002299 complementary DNA Substances 0.000 description 4
- 230000002950 deficient Effects 0.000 description 4
- 229940121647 egfr inhibitor Drugs 0.000 description 4
- 229960001433 erlotinib Drugs 0.000 description 4
- 238000009396 hybridization Methods 0.000 description 4
- KTUFNOKKBVMGRW-UHFFFAOYSA-N imatinib Chemical compound C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 KTUFNOKKBVMGRW-UHFFFAOYSA-N 0.000 description 4
- 210000002865 immune cell Anatomy 0.000 description 4
- 238000001727 in vivo Methods 0.000 description 4
- 208000015181 infectious disease Diseases 0.000 description 4
- 229940047122 interleukins Drugs 0.000 description 4
- 238000001990 intravenous administration Methods 0.000 description 4
- BCFGMOOMADDAQU-UHFFFAOYSA-N lapatinib Chemical compound O1C(CNCCS(=O)(=O)C)=CC=C1C1=CC=C(N=CN=C2NC=3C=C(Cl)C(OCC=4C=C(F)C=CC=4)=CC=3)C2=C1 BCFGMOOMADDAQU-UHFFFAOYSA-N 0.000 description 4
- 239000007788 liquid Substances 0.000 description 4
- 210000004698 lymphocyte Anatomy 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 230000000869 mutational effect Effects 0.000 description 4
- 238000011275 oncology therapy Methods 0.000 description 4
- 230000001575 pathological effect Effects 0.000 description 4
- 239000008194 pharmaceutical composition Substances 0.000 description 4
- 239000000843 powder Substances 0.000 description 4
- 102000004196 processed proteins & peptides Human genes 0.000 description 4
- 230000011664 signaling Effects 0.000 description 4
- 239000002904 solvent Substances 0.000 description 4
- 239000000758 substrate Substances 0.000 description 4
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 4
- 238000003786 synthesis reaction Methods 0.000 description 4
- WYWHKKSPHMUBEB-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 4
- 229960000241 vandetanib Drugs 0.000 description 4
- UHTHHESEBZOYNR-UHFFFAOYSA-N vandetanib Chemical compound COC1=CC(C(/N=CN2)=N/C=3C(=CC(Br)=CC=3)F)=C2C=C1OCC1CCN(C)CC1 UHTHHESEBZOYNR-UHFFFAOYSA-N 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- RTQWWZBSTRGEAV-PKHIMPSTSA-N 2-[[(2s)-2-[bis(carboxymethyl)amino]-3-[4-(methylcarbamoylamino)phenyl]propyl]-[2-[bis(carboxymethyl)amino]propyl]amino]acetic acid Chemical compound CNC(=O)NC1=CC=C(C[C@@H](CN(CC(C)N(CC(O)=O)CC(O)=O)CC(O)=O)N(CC(O)=O)CC(O)=O)C=C1 RTQWWZBSTRGEAV-PKHIMPSTSA-N 0.000 description 3
- 108020005544 Antisense RNA Proteins 0.000 description 3
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 108010008177 Fd immunoglobulins Proteins 0.000 description 3
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 3
- 101000884271 Homo sapiens Signal transducer CD24 Proteins 0.000 description 3
- 101000818543 Homo sapiens Tyrosine-protein kinase ZAP-70 Proteins 0.000 description 3
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 3
- 108010047761 Interferon-alpha Proteins 0.000 description 3
- 102000006992 Interferon-alpha Human genes 0.000 description 3
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 3
- 239000005517 L01XE01 - Imatinib Substances 0.000 description 3
- 239000002147 L01XE04 - Sunitinib Substances 0.000 description 3
- 239000002136 L01XE07 - Lapatinib Substances 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 3
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 3
- 108090000873 Receptor Protein-Tyrosine Kinases Proteins 0.000 description 3
- 102000004278 Receptor Protein-Tyrosine Kinases Human genes 0.000 description 3
- 102100038081 Signal transducer CD24 Human genes 0.000 description 3
- 108010078814 Tumor Suppressor Protein p53 Proteins 0.000 description 3
- 102100021125 Tyrosine-protein kinase ZAP-70 Human genes 0.000 description 3
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 3
- 239000003242 anti bacterial agent Substances 0.000 description 3
- 230000001640 apoptogenic effect Effects 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 229930195731 calicheamicin Natural products 0.000 description 3
- HXCHCVDVKSCDHU-LULTVBGHSA-N calicheamicin Chemical compound C1[C@H](OC)[C@@H](NCC)CO[C@H]1O[C@H]1[C@H](O[C@@H]2C\3=C(NC(=O)OC)C(=O)C[C@](C/3=C/CSSSC)(O)C#C\C=C/C#C2)O[C@H](C)[C@@H](NO[C@@H]2O[C@H](C)[C@@H](SC(=O)C=3C(=C(OC)C(O[C@H]4[C@@H]([C@H](OC)[C@@H](O)[C@H](C)O4)O)=C(I)C=3C)OC)[C@@H](O)C2)[C@@H]1O HXCHCVDVKSCDHU-LULTVBGHSA-N 0.000 description 3
- 230000015556 catabolic process Effects 0.000 description 3
- 230000010261 cell growth Effects 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 238000012512 characterization method Methods 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 230000000295 complement effect Effects 0.000 description 3
- 239000003184 complementary RNA Substances 0.000 description 3
- 230000007423 decrease Effects 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 238000006731 degradation reaction Methods 0.000 description 3
- 238000003745 diagnosis Methods 0.000 description 3
- 230000003828 downregulation Effects 0.000 description 3
- 239000012636 effector Substances 0.000 description 3
- 238000011156 evaluation Methods 0.000 description 3
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 3
- 239000007850 fluorescent dye Substances 0.000 description 3
- 229960002949 fluorouracil Drugs 0.000 description 3
- XGALLCVXEZPNRQ-UHFFFAOYSA-N gefitinib Chemical compound C=12C=C(OCCCN3CCOCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 XGALLCVXEZPNRQ-UHFFFAOYSA-N 0.000 description 3
- 239000003102 growth factor Substances 0.000 description 3
- 210000004408 hybridoma Anatomy 0.000 description 3
- 229960001001 ibritumomab tiuxetan Drugs 0.000 description 3
- 229960002411 imatinib Drugs 0.000 description 3
- 230000002519 immonomodulatory effect Effects 0.000 description 3
- 230000001900 immune effect Effects 0.000 description 3
- 238000003364 immunohistochemistry Methods 0.000 description 3
- 230000001024 immunotherapeutic effect Effects 0.000 description 3
- 238000011534 incubation Methods 0.000 description 3
- 239000004615 ingredient Substances 0.000 description 3
- 230000003993 interaction Effects 0.000 description 3
- 230000031261 interleukin-10 production Effects 0.000 description 3
- 238000007912 intraperitoneal administration Methods 0.000 description 3
- 150000002500 ions Chemical group 0.000 description 3
- 229960004891 lapatinib Drugs 0.000 description 3
- 230000036210 malignancy Effects 0.000 description 3
- 230000003211 malignant effect Effects 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 210000003519 mature b lymphocyte Anatomy 0.000 description 3
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 3
- 229960000485 methotrexate Drugs 0.000 description 3
- 244000005700 microbiome Species 0.000 description 3
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 3
- OHDXDNUPVVYWOV-UHFFFAOYSA-N n-methyl-1-(2-naphthalen-1-ylsulfanylphenyl)methanamine Chemical compound CNCC1=CC=CC=C1SC1=CC=CC2=CC=CC=C12 OHDXDNUPVVYWOV-UHFFFAOYSA-N 0.000 description 3
- 210000000822 natural killer cell Anatomy 0.000 description 3
- 239000002773 nucleotide Substances 0.000 description 3
- 125000003729 nucleotide group Chemical group 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- PHEDXBVPIONUQT-RGYGYFBISA-N phorbol 13-acetate 12-myristate Chemical compound C([C@]1(O)C(=O)C(C)=C[C@H]1[C@@]1(O)[C@H](C)[C@H]2OC(=O)CCCCCCCCCCCCC)C(CO)=C[C@H]1[C@H]1[C@]2(OC(C)=O)C1(C)C PHEDXBVPIONUQT-RGYGYFBISA-N 0.000 description 3
- 239000013600 plasmid vector Substances 0.000 description 3
- 229920001223 polyethylene glycol Polymers 0.000 description 3
- 229920001184 polypeptide Polymers 0.000 description 3
- 238000003753 real-time PCR Methods 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 230000009711 regulatory function Effects 0.000 description 3
- 229960004641 rituximab Drugs 0.000 description 3
- 238000012216 screening Methods 0.000 description 3
- 210000005212 secondary lymphoid organ Anatomy 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- 230000004936 stimulating effect Effects 0.000 description 3
- WINHZLLDWRZWRT-ATVHPVEESA-N sunitinib Chemical compound CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C WINHZLLDWRZWRT-ATVHPVEESA-N 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- 229960005267 tositumomab Drugs 0.000 description 3
- 230000002103 transcriptional effect Effects 0.000 description 3
- 238000010361 transduction Methods 0.000 description 3
- 230000026683 transduction Effects 0.000 description 3
- 230000014616 translation Effects 0.000 description 3
- 239000013603 viral vector Substances 0.000 description 3
- 230000003612 virological effect Effects 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 2
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 2
- 108010032595 Antibody Binding Sites Proteins 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 2
- 239000005461 Canertinib Substances 0.000 description 2
- GAGWJHPBXLXJQN-UORFTKCHSA-N Capecitabine Chemical compound C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](C)O1 GAGWJHPBXLXJQN-UORFTKCHSA-N 0.000 description 2
- 229940046168 CpG oligodeoxynucleotide Drugs 0.000 description 2
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 description 2
- 108010092160 Dactinomycin Proteins 0.000 description 2
- ZBNZXTGUTAYRHI-UHFFFAOYSA-N Dasatinib Chemical compound C=1C(N2CCN(CCO)CC2)=NC(C)=NC=1NC(S1)=NC=C1C(=O)NC1=C(C)C=CC=C1Cl ZBNZXTGUTAYRHI-UHFFFAOYSA-N 0.000 description 2
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 2
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 2
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 2
- 102100026720 Interferon beta Human genes 0.000 description 2
- 108090000467 Interferon-beta Proteins 0.000 description 2
- 102000004551 Interleukin-10 Receptors Human genes 0.000 description 2
- 108010017550 Interleukin-10 Receptors Proteins 0.000 description 2
- 239000005411 L01XE02 - Gefitinib Substances 0.000 description 2
- 239000002118 L01XE12 - Vandetanib Substances 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 238000000585 Mann–Whitney U test Methods 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 2
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 2
- 239000000020 Nitrocellulose Substances 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 241000235648 Pichia Species 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 2
- 108091028664 Ribonucleotide Proteins 0.000 description 2
- 108010017324 STAT3 Transcription Factor Proteins 0.000 description 2
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 2
- 102100024040 Signal transducer and activator of transcription 3 Human genes 0.000 description 2
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 2
- 108020004459 Small interfering RNA Proteins 0.000 description 2
- 230000005867 T cell response Effects 0.000 description 2
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 2
- FOCVUCIESVLUNU-UHFFFAOYSA-N Thiotepa Chemical compound C1CN1P(N1CC1)(=S)N1CC1 FOCVUCIESVLUNU-UHFFFAOYSA-N 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 108091008605 VEGF receptors Proteins 0.000 description 2
- 102000009484 Vascular Endothelial Growth Factor Receptors Human genes 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- 229940100198 alkylating agent Drugs 0.000 description 2
- 239000002168 alkylating agent Substances 0.000 description 2
- 239000012491 analyte Substances 0.000 description 2
- 230000033115 angiogenesis Effects 0.000 description 2
- 230000000340 anti-metabolite Effects 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 229940100197 antimetabolite Drugs 0.000 description 2
- 239000002256 antimetabolite Substances 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 210000000649 b-lymphocyte subset Anatomy 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 238000001815 biotherapy Methods 0.000 description 2
- 229940022399 cancer vaccine Drugs 0.000 description 2
- 238000009566 cancer vaccine Methods 0.000 description 2
- 229950002826 canertinib Drugs 0.000 description 2
- OMZCMEYTWSXEPZ-UHFFFAOYSA-N canertinib Chemical compound C1=C(Cl)C(F)=CC=C1NC1=NC=NC2=CC(OCCCN3CCOCC3)=C(NC(=O)C=C)C=C12 OMZCMEYTWSXEPZ-UHFFFAOYSA-N 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 108091092356 cellular DNA Proteins 0.000 description 2
- 229960005395 cetuximab Drugs 0.000 description 2
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 2
- 229960004630 chlorambucil Drugs 0.000 description 2
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- 230000004186 co-expression Effects 0.000 description 2
- 239000012228 culture supernatant Substances 0.000 description 2
- 229960002465 dabrafenib Drugs 0.000 description 2
- BFSMGDJOXZAERB-UHFFFAOYSA-N dabrafenib Chemical compound S1C(C(C)(C)C)=NC(C=2C(=C(NS(=O)(=O)C=3C(=CC=CC=3F)F)C=CC=2)F)=C1C1=CC=NC(N)=N1 BFSMGDJOXZAERB-UHFFFAOYSA-N 0.000 description 2
- 229960003901 dacarbazine Drugs 0.000 description 2
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 2
- 230000006735 deficit Effects 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 230000000779 depleting effect Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 230000004069 differentiation Effects 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 239000002612 dispersion medium Substances 0.000 description 2
- 230000002616 endonucleolytic effect Effects 0.000 description 2
- OVBPIULPVIDEAO-LBPRGKRZSA-N folic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-LBPRGKRZSA-N 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- CHPZKNULDCNCBW-UHFFFAOYSA-N gallium nitrate Chemical compound [Ga+3].[O-][N+]([O-])=O.[O-][N+]([O-])=O.[O-][N+]([O-])=O CHPZKNULDCNCBW-UHFFFAOYSA-N 0.000 description 2
- 229960002584 gefitinib Drugs 0.000 description 2
- 229960003297 gemtuzumab ozogamicin Drugs 0.000 description 2
- 230000009368 gene silencing by RNA Effects 0.000 description 2
- 238000001415 gene therapy Methods 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 210000002443 helper t lymphocyte Anatomy 0.000 description 2
- 229960001101 ifosfamide Drugs 0.000 description 2
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 2
- 230000037451 immune surveillance Effects 0.000 description 2
- 229940127121 immunoconjugate Drugs 0.000 description 2
- 229940072221 immunoglobulins Drugs 0.000 description 2
- 239000000568 immunological adjuvant Substances 0.000 description 2
- 230000008975 immunomodulatory function Effects 0.000 description 2
- 238000001114 immunoprecipitation Methods 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 102000006639 indoleamine 2,3-dioxygenase Human genes 0.000 description 2
- 108020004201 indoleamine 2,3-dioxygenase Proteins 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 230000037431 insertion Effects 0.000 description 2
- 230000010354 integration Effects 0.000 description 2
- PGHMRUGBZOYCAA-UHFFFAOYSA-N ionomycin Natural products O1C(CC(O)C(C)C(O)C(C)C=CCC(C)CC(C)C(O)=CC(=O)C(C)CC(C)CC(CCC(O)=O)C)CCC1(C)C1OC(C)(C(C)O)CC1 PGHMRUGBZOYCAA-UHFFFAOYSA-N 0.000 description 2
- PGHMRUGBZOYCAA-ADZNBVRBSA-N ionomycin Chemical compound O1[C@H](C[C@H](O)[C@H](C)[C@H](O)[C@H](C)/C=C/C[C@@H](C)C[C@@H](C)C(/O)=C/C(=O)[C@@H](C)C[C@@H](C)C[C@@H](CCC(O)=O)C)CC[C@@]1(C)[C@@H]1O[C@](C)([C@@H](C)O)CC1 PGHMRUGBZOYCAA-ADZNBVRBSA-N 0.000 description 2
- VHOGYURTWQBHIL-UHFFFAOYSA-N leflunomide Chemical compound O1N=CC(C(=O)NC=2C=CC(=CC=2)C(F)(F)F)=C1C VHOGYURTWQBHIL-UHFFFAOYSA-N 0.000 description 2
- 208000032839 leukemia Diseases 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 229960001428 mercaptopurine Drugs 0.000 description 2
- 230000002503 metabolic effect Effects 0.000 description 2
- 230000011987 methylation Effects 0.000 description 2
- 238000007069 methylation reaction Methods 0.000 description 2
- 239000004005 microsphere Substances 0.000 description 2
- 229960001156 mitoxantrone Drugs 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- QZGIWPZCWHMVQL-UIYAJPBUSA-N neocarzinostatin chromophore Chemical compound O1[C@H](C)[C@H](O)[C@H](O)[C@@H](NC)[C@H]1O[C@@H]1C/2=C/C#C[C@H]3O[C@@]3([C@@H]3OC(=O)OC3)C#CC\2=C[C@H]1OC(=O)C1=C(O)C=CC2=C(C)C=C(OC)C=C12 QZGIWPZCWHMVQL-UIYAJPBUSA-N 0.000 description 2
- 229920001220 nitrocellulos Polymers 0.000 description 2
- 229960002450 ofatumumab Drugs 0.000 description 2
- 239000003921 oil Substances 0.000 description 2
- 235000019198 oils Nutrition 0.000 description 2
- 229940127084 other anti-cancer agent Drugs 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 238000004091 panning Methods 0.000 description 2
- 238000002823 phage display Methods 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 239000012071 phase Substances 0.000 description 2
- 210000003720 plasmablast Anatomy 0.000 description 2
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 2
- 239000001397 quillaja saponaria molina bark Substances 0.000 description 2
- 238000003127 radioimmunoassay Methods 0.000 description 2
- 238000001959 radiotherapy Methods 0.000 description 2
- 229960004622 raloxifene Drugs 0.000 description 2
- GZUITABIAKMVPG-UHFFFAOYSA-N raloxifene Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCCC3)=CC=2)C2=CC=C(O)C=C2S1 GZUITABIAKMVPG-UHFFFAOYSA-N 0.000 description 2
- 238000007634 remodeling Methods 0.000 description 2
- 239000002336 ribonucleotide Substances 0.000 description 2
- 125000002652 ribonucleotide group Chemical group 0.000 description 2
- 229930182490 saponin Natural products 0.000 description 2
- 150000007949 saponins Chemical class 0.000 description 2
- 230000003248 secreting effect Effects 0.000 description 2
- WUWDLXZGHZSWQZ-WQLSENKSSA-N semaxanib Chemical compound N1C(C)=CC(C)=C1\C=C/1C2=CC=CC=C2NC\1=O WUWDLXZGHZSWQZ-WQLSENKSSA-N 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 239000001488 sodium phosphate Substances 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 238000003860 storage Methods 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 229960001796 sunitinib Drugs 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 238000002626 targeted therapy Methods 0.000 description 2
- 229940124597 therapeutic agent Drugs 0.000 description 2
- 229940126585 therapeutic drug Drugs 0.000 description 2
- 229960001196 thiotepa Drugs 0.000 description 2
- 229960003087 tioguanine Drugs 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- 238000013519 translation Methods 0.000 description 2
- 230000001960 triggered effect Effects 0.000 description 2
- GETQZCLCWQTVFV-UHFFFAOYSA-N trimethylamine Chemical compound CN(C)C GETQZCLCWQTVFV-UHFFFAOYSA-N 0.000 description 2
- 241000701161 unidentified adenovirus Species 0.000 description 2
- 230000003827 upregulation Effects 0.000 description 2
- 238000010200 validation analysis Methods 0.000 description 2
- 229950000578 vatalanib Drugs 0.000 description 2
- YCOYDOIWSSHVCK-UHFFFAOYSA-N vatalanib Chemical compound C1=CC(Cl)=CC=C1NC(C1=CC=CC=C11)=NN=C1CC1=CC=NC=C1 YCOYDOIWSSHVCK-UHFFFAOYSA-N 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- 230000003442 weekly effect Effects 0.000 description 2
- NNJPGOLRFBJNIW-HNNXBMFYSA-N (-)-demecolcine Chemical compound C1=C(OC)C(=O)C=C2[C@@H](NC)CCC3=CC(OC)=C(OC)C(OC)=C3C2=C1 NNJPGOLRFBJNIW-HNNXBMFYSA-N 0.000 description 1
- WCWUXEGQKLTGDX-LLVKDONJSA-N (2R)-1-[[4-[(4-fluoro-2-methyl-1H-indol-5-yl)oxy]-5-methyl-6-pyrrolo[2,1-f][1,2,4]triazinyl]oxy]-2-propanol Chemical compound C1=C2NC(C)=CC2=C(F)C(OC2=NC=NN3C=C(C(=C32)C)OC[C@H](O)C)=C1 WCWUXEGQKLTGDX-LLVKDONJSA-N 0.000 description 1
- WDQLRUYAYXDIFW-RWKIJVEZSA-N (2r,3r,4s,5r,6r)-4-[(2s,3r,4s,5r,6r)-3,5-dihydroxy-4-[(2r,3r,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-6-[[(2r,3r,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxymethyl]oxan-2-yl]oxy-6-(hydroxymethyl)oxane-2,3,5-triol Chemical compound O[C@@H]1[C@@H](CO)O[C@@H](O)[C@H](O)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)O)[C@H](O)[C@@H](CO[C@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)O)O1 WDQLRUYAYXDIFW-RWKIJVEZSA-N 0.000 description 1
- FLWWDYNPWOSLEO-HQVZTVAUSA-N (2s)-2-[[4-[1-(2-amino-4-oxo-1h-pteridin-6-yl)ethyl-methylamino]benzoyl]amino]pentanedioic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1C(C)N(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FLWWDYNPWOSLEO-HQVZTVAUSA-N 0.000 description 1
- CGMTUJFWROPELF-YPAAEMCBSA-N (3E,5S)-5-[(2S)-butan-2-yl]-3-(1-hydroxyethylidene)pyrrolidine-2,4-dione Chemical compound CC[C@H](C)[C@@H]1NC(=O)\C(=C(/C)O)C1=O CGMTUJFWROPELF-YPAAEMCBSA-N 0.000 description 1
- KCOYQXZDFIIGCY-CZIZESTLSA-N (3e)-4-amino-5-fluoro-3-[5-(4-methylpiperazin-1-yl)-1,3-dihydrobenzimidazol-2-ylidene]quinolin-2-one Chemical compound C1CN(C)CCN1C1=CC=C(N\C(N2)=C/3C(=C4C(F)=CC=CC4=NC\3=O)N)C2=C1 KCOYQXZDFIIGCY-CZIZESTLSA-N 0.000 description 1
- TVIRNGFXQVMMGB-OFWIHYRESA-N (3s,6r,10r,13e,16s)-16-[(2r,3r,4s)-4-chloro-3-hydroxy-4-phenylbutan-2-yl]-10-[(3-chloro-4-methoxyphenyl)methyl]-6-methyl-3-(2-methylpropyl)-1,4-dioxa-8,11-diazacyclohexadec-13-ene-2,5,9,12-tetrone Chemical compound C1=C(Cl)C(OC)=CC=C1C[C@@H]1C(=O)NC[C@@H](C)C(=O)O[C@@H](CC(C)C)C(=O)O[C@H]([C@H](C)[C@@H](O)[C@@H](Cl)C=2C=CC=CC=2)C/C=C/C(=O)N1 TVIRNGFXQVMMGB-OFWIHYRESA-N 0.000 description 1
- XRBSKUSTLXISAB-XVVDYKMHSA-N (5r,6r,7r,8r)-8-hydroxy-7-(hydroxymethyl)-5-(3,4,5-trimethoxyphenyl)-5,6,7,8-tetrahydrobenzo[f][1,3]benzodioxole-6-carboxylic acid Chemical compound COC1=C(OC)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@H](O)[C@@H](CO)[C@@H]2C(O)=O)=C1 XRBSKUSTLXISAB-XVVDYKMHSA-N 0.000 description 1
- XRBSKUSTLXISAB-UHFFFAOYSA-N (7R,7'R,8R,8'R)-form-Podophyllic acid Natural products COC1=C(OC)C(OC)=CC(C2C3=CC=4OCOC=4C=C3C(O)C(CO)C2C(O)=O)=C1 XRBSKUSTLXISAB-UHFFFAOYSA-N 0.000 description 1
- AESVUZLWRXEGEX-DKCAWCKPSA-N (7S,9R)-7-[(2S,4R,5R,6R)-4-amino-5-hydroxy-6-methyloxan-2-yl]oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7H-tetracene-5,12-dione iron(3+) Chemical compound [Fe+3].COc1cccc2C(=O)c3c(O)c4C[C@@](O)(C[C@H](O[C@@H]5C[C@@H](N)[C@@H](O)[C@@H](C)O5)c4c(O)c3C(=O)c12)C(=O)CO AESVUZLWRXEGEX-DKCAWCKPSA-N 0.000 description 1
- JXVAMODRWBNUSF-KZQKBALLSA-N (7s,9r,10r)-7-[(2r,4s,5s,6s)-5-[[(2s,4as,5as,7s,9s,9ar,10ar)-2,9-dimethyl-3-oxo-4,4a,5a,6,7,9,9a,10a-octahydrodipyrano[4,2-a:4',3'-e][1,4]dioxin-7-yl]oxy]-4-(dimethylamino)-6-methyloxan-2-yl]oxy-10-[(2s,4s,5s,6s)-4-(dimethylamino)-5-hydroxy-6-methyloxan-2 Chemical compound O([C@@H]1C2=C(O)C=3C(=O)C4=CC=CC(O)=C4C(=O)C=3C(O)=C2[C@@H](O[C@@H]2O[C@@H](C)[C@@H](O[C@@H]3O[C@@H](C)[C@H]4O[C@@H]5O[C@@H](C)C(=O)C[C@@H]5O[C@H]4C3)[C@H](C2)N(C)C)C[C@]1(O)CC)[C@H]1C[C@H](N(C)C)[C@H](O)[C@H](C)O1 JXVAMODRWBNUSF-KZQKBALLSA-N 0.000 description 1
- INAUWOVKEZHHDM-PEDBPRJASA-N (7s,9s)-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-7-[(2r,4s,5s,6s)-5-hydroxy-6-methyl-4-morpholin-4-yloxan-2-yl]oxy-4-methoxy-8,10-dihydro-7h-tetracene-5,12-dione;hydrochloride Chemical compound Cl.N1([C@H]2C[C@@H](O[C@@H](C)[C@H]2O)O[C@H]2C[C@@](O)(CC=3C(O)=C4C(=O)C=5C=CC=C(C=5C(=O)C4=C(O)C=32)OC)C(=O)CO)CCOCC1 INAUWOVKEZHHDM-PEDBPRJASA-N 0.000 description 1
- RCFNNLSZHVHCEK-IMHLAKCZSA-N (7s,9s)-7-(4-amino-6-methyloxan-2-yl)oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7h-tetracene-5,12-dione;hydrochloride Chemical compound [Cl-].O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)C1CC([NH3+])CC(C)O1 RCFNNLSZHVHCEK-IMHLAKCZSA-N 0.000 description 1
- NOPNWHSMQOXAEI-PUCKCBAPSA-N (7s,9s)-7-[(2r,4s,5s,6s)-4-(2,3-dihydropyrrol-1-yl)-5-hydroxy-6-methyloxan-2-yl]oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7h-tetracene-5,12-dione Chemical compound N1([C@H]2C[C@@H](O[C@@H](C)[C@H]2O)O[C@H]2C[C@@](O)(CC=3C(O)=C4C(=O)C=5C=CC=C(C=5C(=O)C4=C(O)C=32)OC)C(=O)CO)CCC=C1 NOPNWHSMQOXAEI-PUCKCBAPSA-N 0.000 description 1
- FPVKHBSQESCIEP-UHFFFAOYSA-N (8S)-3-(2-deoxy-beta-D-erythro-pentofuranosyl)-3,6,7,8-tetrahydroimidazo[4,5-d][1,3]diazepin-8-ol Natural products C1C(O)C(CO)OC1N1C(NC=NCC2O)=C2N=C1 FPVKHBSQESCIEP-UHFFFAOYSA-N 0.000 description 1
- IEXUMDBQLIVNHZ-YOUGDJEHSA-N (8s,11r,13r,14s,17s)-11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(3-hydroxypropyl)-13-methyl-1,2,6,7,8,11,12,14,15,16-decahydrocyclopenta[a]phenanthren-3-one Chemical compound C1=CC(N(C)C)=CC=C1[C@@H]1C2=C3CCC(=O)C=C3CC[C@H]2[C@H](CC[C@]2(O)CCCO)[C@@]2(C)C1 IEXUMDBQLIVNHZ-YOUGDJEHSA-N 0.000 description 1
- LKJPYSCBVHEWIU-KRWDZBQOSA-N (R)-bicalutamide Chemical compound C([C@@](O)(C)C(=O)NC=1C=C(C(C#N)=CC=1)C(F)(F)F)S(=O)(=O)C1=CC=C(F)C=C1 LKJPYSCBVHEWIU-KRWDZBQOSA-N 0.000 description 1
- AGNGYMCLFWQVGX-AGFFZDDWSA-N (e)-1-[(2s)-2-amino-2-carboxyethoxy]-2-diazonioethenolate Chemical compound OC(=O)[C@@H](N)CO\C([O-])=C\[N+]#N AGNGYMCLFWQVGX-AGFFZDDWSA-N 0.000 description 1
- FONKWHRXTPJODV-DNQXCXABSA-N 1,3-bis[2-[(8s)-8-(chloromethyl)-4-hydroxy-1-methyl-7,8-dihydro-3h-pyrrolo[3,2-e]indole-6-carbonyl]-1h-indol-5-yl]urea Chemical compound C1([C@H](CCl)CN2C(=O)C=3NC4=CC=C(C=C4C=3)NC(=O)NC=3C=C4C=C(NC4=CC=3)C(=O)N3C4=CC(O)=C5NC=C(C5=C4[C@H](CCl)C3)C)=C2C=C(O)C2=C1C(C)=CN2 FONKWHRXTPJODV-DNQXCXABSA-N 0.000 description 1
- VPBYZLCHOKSGRX-UHFFFAOYSA-N 1-[2-chloro-4-(6,7-dimethoxyquinazolin-4-yl)oxyphenyl]-3-propylurea Chemical compound C1=C(Cl)C(NC(=O)NCCC)=CC=C1OC1=NC=NC2=CC(OC)=C(OC)C=C12 VPBYZLCHOKSGRX-UHFFFAOYSA-N 0.000 description 1
- SPMVMDHWKHCIDT-UHFFFAOYSA-N 1-[2-chloro-4-[(6,7-dimethoxy-4-quinolinyl)oxy]phenyl]-3-(5-methyl-3-isoxazolyl)urea Chemical compound C=12C=C(OC)C(OC)=CC2=NC=CC=1OC(C=C1Cl)=CC=C1NC(=O)NC=1C=C(C)ON=1 SPMVMDHWKHCIDT-UHFFFAOYSA-N 0.000 description 1
- YABJJWZLRMPFSI-UHFFFAOYSA-N 1-methyl-5-[[2-[5-(trifluoromethyl)-1H-imidazol-2-yl]-4-pyridinyl]oxy]-N-[4-(trifluoromethyl)phenyl]-2-benzimidazolamine Chemical compound N=1C2=CC(OC=3C=C(N=CC=3)C=3NC(=CN=3)C(F)(F)F)=CC=C2N(C)C=1NC1=CC=C(C(F)(F)F)C=C1 YABJJWZLRMPFSI-UHFFFAOYSA-N 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- 102100026210 1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase gamma-2 Human genes 0.000 description 1
- BTOTXLJHDSNXMW-POYBYMJQSA-N 2,3-dideoxyuridine Chemical compound O1[C@H](CO)CC[C@@H]1N1C(=O)NC(=O)C=C1 BTOTXLJHDSNXMW-POYBYMJQSA-N 0.000 description 1
- BOMZMNZEXMAQQW-UHFFFAOYSA-N 2,5,11-trimethyl-6h-pyrido[4,3-b]carbazol-2-ium-9-ol;acetate Chemical compound CC([O-])=O.C[N+]1=CC=C2C(C)=C(NC=3C4=CC(O)=CC=3)C4=C(C)C2=C1 BOMZMNZEXMAQQW-UHFFFAOYSA-N 0.000 description 1
- QCXJFISCRQIYID-IAEPZHFASA-N 2-amino-1-n-[(3s,6s,7r,10s,16s)-3-[(2s)-butan-2-yl]-7,11,14-trimethyl-2,5,9,12,15-pentaoxo-10-propan-2-yl-8-oxa-1,4,11,14-tetrazabicyclo[14.3.0]nonadecan-6-yl]-4,6-dimethyl-3-oxo-9-n-[(3s,6s,7r,10s,16s)-7,11,14-trimethyl-2,5,9,12,15-pentaoxo-3,10-di(propa Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N=C2C(C(=O)N[C@@H]3C(=O)N[C@H](C(N4CCC[C@H]4C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]3C)=O)[C@@H](C)CC)=C(N)C(=O)C(C)=C2O2)C2=C(C)C=C1 QCXJFISCRQIYID-IAEPZHFASA-N 0.000 description 1
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 1
- VNBAOSVONFJBKP-UHFFFAOYSA-N 2-chloro-n,n-bis(2-chloroethyl)propan-1-amine;hydrochloride Chemical compound Cl.CC(Cl)CN(CCCl)CCCl VNBAOSVONFJBKP-UHFFFAOYSA-N 0.000 description 1
- FJCDSQATIJKQKA-UHFFFAOYSA-N 2-fluoro-n-[[5-(6-methylpyridin-2-yl)-4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-1h-imidazol-2-yl]methyl]aniline Chemical compound CC1=CC=CC(C2=C(N=C(CNC=3C(=CC=CC=3)F)N2)C2=CN3N=CN=C3C=C2)=N1 FJCDSQATIJKQKA-UHFFFAOYSA-N 0.000 description 1
- YIMDLWDNDGKDTJ-QLKYHASDSA-N 3'-deamino-3'-(3-cyanomorpholin-4-yl)doxorubicin Chemical compound N1([C@H]2C[C@@H](O[C@@H](C)[C@H]2O)O[C@H]2C[C@@](O)(CC=3C(O)=C4C(=O)C=5C=CC=C(C=5C(=O)C4=C(O)C=32)OC)C(=O)CO)CCOCC1C#N YIMDLWDNDGKDTJ-QLKYHASDSA-N 0.000 description 1
- NDMPLJNOPCLANR-UHFFFAOYSA-N 3,4-dihydroxy-15-(4-hydroxy-18-methoxycarbonyl-5,18-seco-ibogamin-18-yl)-16-methoxy-1-methyl-6,7-didehydro-aspidospermidine-3-carboxylic acid methyl ester Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 NDMPLJNOPCLANR-UHFFFAOYSA-N 0.000 description 1
- AXRCEOKUDYDWLF-UHFFFAOYSA-N 3-(1-methyl-3-indolyl)-4-[1-[1-(2-pyridinylmethyl)-4-piperidinyl]-3-indolyl]pyrrole-2,5-dione Chemical compound C12=CC=CC=C2N(C)C=C1C(C(NC1=O)=O)=C1C(C1=CC=CC=C11)=CN1C(CC1)CCN1CC1=CC=CC=N1 AXRCEOKUDYDWLF-UHFFFAOYSA-N 0.000 description 1
- FGTCROZDHDSNIO-UHFFFAOYSA-N 3-(4-quinolinylmethylamino)-N-[4-(trifluoromethoxy)phenyl]-2-thiophenecarboxamide Chemical compound C1=CC(OC(F)(F)F)=CC=C1NC(=O)C1=C(NCC=2C3=CC=CC=C3N=CC=2)C=CS1 FGTCROZDHDSNIO-UHFFFAOYSA-N 0.000 description 1
- AOJJSUZBOXZQNB-VTZDEGQISA-N 4'-epidoxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-VTZDEGQISA-N 0.000 description 1
- XXJWYDDUDKYVKI-UHFFFAOYSA-N 4-[(4-fluoro-2-methyl-1H-indol-5-yl)oxy]-6-methoxy-7-[3-(1-pyrrolidinyl)propoxy]quinazoline Chemical compound COC1=CC2=C(OC=3C(=C4C=C(C)NC4=CC=3)F)N=CN=C2C=C1OCCCN1CCCC1 XXJWYDDUDKYVKI-UHFFFAOYSA-N 0.000 description 1
- DODQJNMQWMSYGS-QPLCGJKRSA-N 4-[(z)-1-[4-[2-(dimethylamino)ethoxy]phenyl]-1-phenylbut-1-en-2-yl]phenol Chemical compound C=1C=C(O)C=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 DODQJNMQWMSYGS-QPLCGJKRSA-N 0.000 description 1
- QFCXANHHBCGMAS-UHFFFAOYSA-N 4-[[4-(4-chloroanilino)furo[2,3-d]pyridazin-7-yl]oxymethyl]-n-methylpyridine-2-carboxamide Chemical compound C1=NC(C(=O)NC)=CC(COC=2C=3OC=CC=3C(NC=3C=CC(Cl)=CC=3)=NN=2)=C1 QFCXANHHBCGMAS-UHFFFAOYSA-N 0.000 description 1
- HHFBDROWDBDFBR-UHFFFAOYSA-N 4-[[9-chloro-7-(2,6-difluorophenyl)-5H-pyrimido[5,4-d][2]benzazepin-2-yl]amino]benzoic acid Chemical compound C1=CC(C(=O)O)=CC=C1NC1=NC=C(CN=C(C=2C3=CC=C(Cl)C=2)C=2C(=CC=CC=2F)F)C3=N1 HHFBDROWDBDFBR-UHFFFAOYSA-N 0.000 description 1
- TVZGACDUOSZQKY-LBPRGKRZSA-N 4-aminofolic acid Chemical compound C1=NC2=NC(N)=NC(N)=C2N=C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 TVZGACDUOSZQKY-LBPRGKRZSA-N 0.000 description 1
- 102100023216 40S ribosomal protein S15 Human genes 0.000 description 1
- 101710092702 47 kDa protein Proteins 0.000 description 1
- IDPUKCWIGUEADI-UHFFFAOYSA-N 5-[bis(2-chloroethyl)amino]uracil Chemical compound ClCCN(CCCl)C1=CNC(=O)NC1=O IDPUKCWIGUEADI-UHFFFAOYSA-N 0.000 description 1
- NMUSYJAQQFHJEW-KVTDHHQDSA-N 5-azacytidine Chemical compound O=C1N=C(N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 NMUSYJAQQFHJEW-KVTDHHQDSA-N 0.000 description 1
- WYXSYVWAUAUWLD-SHUUEZRQSA-N 6-azauridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=N1 WYXSYVWAUAUWLD-SHUUEZRQSA-N 0.000 description 1
- 229960005538 6-diazo-5-oxo-L-norleucine Drugs 0.000 description 1
- YCWQAMGASJSUIP-YFKPBYRVSA-N 6-diazo-5-oxo-L-norleucine Chemical compound OC(=O)[C@@H](N)CCC(=O)C=[N+]=[N-] YCWQAMGASJSUIP-YFKPBYRVSA-N 0.000 description 1
- QKDCLUARMDUUKN-XMMPIXPASA-N 6-ethyl-3-[4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]anilino]-5-[(3r)-1-prop-2-enoylpyrrolidin-3-yl]oxypyrazine-2-carboxamide Chemical compound N1=C(O[C@H]2CN(CC2)C(=O)C=C)C(CC)=NC(C(N)=O)=C1NC(C=C1)=CC=C1N(CC1)CCC1N1CCN(C)CC1 QKDCLUARMDUUKN-XMMPIXPASA-N 0.000 description 1
- UFGQWTWQNIGAEB-UHFFFAOYSA-N 7-chloroquinoline-3-carboxylic acid Chemical compound C1=C(Cl)C=CC2=CC(C(=O)O)=CN=C21 UFGQWTWQNIGAEB-UHFFFAOYSA-N 0.000 description 1
- ZGXJTSGNIOSYLO-UHFFFAOYSA-N 88755TAZ87 Chemical compound NCC(=O)CCC(O)=O ZGXJTSGNIOSYLO-UHFFFAOYSA-N 0.000 description 1
- HDZZVAMISRMYHH-UHFFFAOYSA-N 9beta-Ribofuranosyl-7-deazaadenin Natural products C1=CC=2C(N)=NC=NC=2N1C1OC(CO)C(O)C1O HDZZVAMISRMYHH-UHFFFAOYSA-N 0.000 description 1
- 230000005730 ADP ribosylation Effects 0.000 description 1
- OONFNUWBHFSNBT-HXUWFJFHSA-N AEE788 Chemical compound C1CN(CC)CCN1CC1=CC=C(C=2NC3=NC=NC(N[C@H](C)C=4C=CC=CC=4)=C3C=2)C=C1 OONFNUWBHFSNBT-HXUWFJFHSA-N 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 1
- CEIZFXOZIQNICU-UHFFFAOYSA-N Alternaria alternata Crofton-weed toxin Natural products CCC(C)C1NC(=O)C(C(C)=O)=C1O CEIZFXOZIQNICU-UHFFFAOYSA-N 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 244000303258 Annona diversifolia Species 0.000 description 1
- 235000002198 Annona diversifolia Nutrition 0.000 description 1
- 108010083359 Antigen Receptors Proteins 0.000 description 1
- 102000006306 Antigen Receptors Human genes 0.000 description 1
- 108020004491 Antisense DNA Proteins 0.000 description 1
- 101000719121 Arabidopsis thaliana Protein MEI2-like 1 Proteins 0.000 description 1
- 101100485276 Arabidopsis thaliana XPO1 gene Proteins 0.000 description 1
- 102000014654 Aromatase Human genes 0.000 description 1
- 108010078554 Aromatase Proteins 0.000 description 1
- 241000228212 Aspergillus Species 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- NOWKCMXCCJGMRR-UHFFFAOYSA-N Aziridine Chemical class C1CN1 NOWKCMXCCJGMRR-UHFFFAOYSA-N 0.000 description 1
- 230000003844 B-cell-activation Effects 0.000 description 1
- MLDQJTXFUGDVEO-UHFFFAOYSA-N BAY-43-9006 Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=CC(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 MLDQJTXFUGDVEO-UHFFFAOYSA-N 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 108700003785 Baculoviral IAP Repeat-Containing 3 Proteins 0.000 description 1
- 102100021662 Baculoviral IAP repeat-containing protein 3 Human genes 0.000 description 1
- VGGGPCQERPFHOB-MCIONIFRSA-N Bestatin Chemical compound CC(C)C[C@H](C(O)=O)NC(=O)[C@@H](O)[C@H](N)CC1=CC=CC=C1 VGGGPCQERPFHOB-MCIONIFRSA-N 0.000 description 1
- 101150104237 Birc3 gene Proteins 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- MBABCNBNDNGODA-LTGLSHGVSA-N Bullatacin Natural products O=C1C(C[C@H](O)CCCCCCCCCC[C@@H](O)[C@@H]2O[C@@H]([C@@H]3O[C@H]([C@@H](O)CCCCCCCCCC)CC3)CC2)=C[C@H](C)O1 MBABCNBNDNGODA-LTGLSHGVSA-N 0.000 description 1
- KGGVWMAPBXIMEM-ZRTAFWODSA-N Bullatacinone Chemical compound O1[C@@H]([C@@H](O)CCCCCCCCCC)CC[C@@H]1[C@@H]1O[C@@H]([C@H](O)CCCCCCCCCC[C@H]2OC(=O)[C@H](CC(C)=O)C2)CC1 KGGVWMAPBXIMEM-ZRTAFWODSA-N 0.000 description 1
- KGGVWMAPBXIMEM-JQFCFGFHSA-N Bullatacinone Natural products O=C(C[C@H]1C(=O)O[C@H](CCCCCCCCCC[C@H](O)[C@@H]2O[C@@H]([C@@H]3O[C@@H]([C@@H](O)CCCCCCCCCC)CC3)CC2)C1)C KGGVWMAPBXIMEM-JQFCFGFHSA-N 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- 229940124292 CD20 monoclonal antibody Drugs 0.000 description 1
- LLVZBTWPGQVVLW-SNAWJCMRSA-N CP-724714 Chemical compound C12=CC(/C=C/CNC(=O)COC)=CC=C2N=CN=C1NC(C=C1C)=CC=C1OC1=CC=C(C)N=C1 LLVZBTWPGQVVLW-SNAWJCMRSA-N 0.000 description 1
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- KLWPJMFMVPTNCC-UHFFFAOYSA-N Camptothecin Natural products CCC1(O)C(=O)OCC2=C1C=C3C4Nc5ccccc5C=C4CN3C2=O KLWPJMFMVPTNCC-UHFFFAOYSA-N 0.000 description 1
- GAGWJHPBXLXJQN-UHFFFAOYSA-N Capecitabine Natural products C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1C1C(O)C(O)C(C)O1 GAGWJHPBXLXJQN-UHFFFAOYSA-N 0.000 description 1
- SHHKQEUPHAENFK-UHFFFAOYSA-N Carboquone Chemical compound O=C1C(C)=C(N2CC2)C(=O)C(C(COC(N)=O)OC)=C1N1CC1 SHHKQEUPHAENFK-UHFFFAOYSA-N 0.000 description 1
- AOCCBINRVIKJHY-UHFFFAOYSA-N Carmofur Chemical compound CCCCCCNC(=O)N1C=C(F)C(=O)NC1=O AOCCBINRVIKJHY-UHFFFAOYSA-N 0.000 description 1
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 1
- 241001227713 Chiron Species 0.000 description 1
- JWBOIMRXGHLCPP-UHFFFAOYSA-N Chloditan Chemical compound C=1C=CC=C(Cl)C=1C(C(Cl)Cl)C1=CC=C(Cl)C=C1 JWBOIMRXGHLCPP-UHFFFAOYSA-N 0.000 description 1
- MKQWTWSXVILIKJ-LXGUWJNJSA-N Chlorozotocin Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](C=O)NC(=O)N(N=O)CCCl MKQWTWSXVILIKJ-LXGUWJNJSA-N 0.000 description 1
- 241000725101 Clea Species 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 229930188224 Cryptophycin Natural products 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 108010041986 DNA Vaccines Proteins 0.000 description 1
- 229940021995 DNA vaccine Drugs 0.000 description 1
- 241000450599 DNA viruses Species 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- WEAHRLBPCANXCN-UHFFFAOYSA-N Daunomycin Natural products CCC1(O)CC(OC2CC(N)C(O)C(C)O2)c3cc4C(=O)c5c(OC)cccc5C(=O)c4c(O)c3C1 WEAHRLBPCANXCN-UHFFFAOYSA-N 0.000 description 1
- NNJPGOLRFBJNIW-UHFFFAOYSA-N Demecolcine Natural products C1=C(OC)C(=O)C=C2C(NC)CCC3=CC(OC)=C(OC)C(OC)=C3C2=C1 NNJPGOLRFBJNIW-UHFFFAOYSA-N 0.000 description 1
- 108010002156 Depsipeptides Proteins 0.000 description 1
- AUGQEEXBDZWUJY-ZLJUKNTDSA-N Diacetoxyscirpenol Chemical compound C([C@]12[C@]3(C)[C@H](OC(C)=O)[C@@H](O)[C@H]1O[C@@H]1C=C(C)CC[C@@]13COC(=O)C)O2 AUGQEEXBDZWUJY-ZLJUKNTDSA-N 0.000 description 1
- AUGQEEXBDZWUJY-UHFFFAOYSA-N Diacetoxyscirpenol Natural products CC(=O)OCC12CCC(C)=CC1OC1C(O)C(OC(C)=O)C2(C)C11CO1 AUGQEEXBDZWUJY-UHFFFAOYSA-N 0.000 description 1
- 229930193152 Dynemicin Natural products 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 229940122558 EGFR antagonist Drugs 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- AFMYMMXSQGUCBK-UHFFFAOYSA-N Endynamicin A Natural products C1#CC=CC#CC2NC(C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C(O)=C3)=C3C34OC32C(C)C(C(O)=O)=C(OC)C41 AFMYMMXSQGUCBK-UHFFFAOYSA-N 0.000 description 1
- SAMRUMKYXPVKPA-VFKOLLTISA-N Enocitabine Chemical compound O=C1N=C(NC(=O)CCCCCCCCCCCCCCCCCCCCC)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 SAMRUMKYXPVKPA-VFKOLLTISA-N 0.000 description 1
- 241000991587 Enterovirus C Species 0.000 description 1
- HTIJFSOGRVMCQR-UHFFFAOYSA-N Epirubicin Natural products COc1cccc2C(=O)c3c(O)c4CC(O)(CC(OC5CC(N)C(=O)C(C)O5)c4c(O)c3C(=O)c12)C(=O)CO HTIJFSOGRVMCQR-UHFFFAOYSA-N 0.000 description 1
- OBMLHUPNRURLOK-XGRAFVIBSA-N Epitiostanol Chemical compound C1[C@@H]2S[C@@H]2C[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CC[C@H]21 OBMLHUPNRURLOK-XGRAFVIBSA-N 0.000 description 1
- 229930189413 Esperamicin Natural products 0.000 description 1
- 108700039887 Essential Genes Proteins 0.000 description 1
- JOYRKODLDBILNP-UHFFFAOYSA-N Ethyl urethane Chemical compound CCOC(N)=O JOYRKODLDBILNP-UHFFFAOYSA-N 0.000 description 1
- 102100028138 F-box/WD repeat-containing protein 7 Human genes 0.000 description 1
- 238000000729 Fisher's exact test Methods 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 101001070329 Geobacillus stearothermophilus 50S ribosomal protein L18 Proteins 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- BLCLNMBMMGCOAS-URPVMXJPSA-N Goserelin Chemical compound C([C@@H](C(=O)N[C@H](COC(C)(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(N)N)C(=O)N1[C@@H](CCC1)C(=O)NNC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 BLCLNMBMMGCOAS-URPVMXJPSA-N 0.000 description 1
- 108010069236 Goserelin Proteins 0.000 description 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 1
- 241000713858 Harvey murine sarcoma virus Species 0.000 description 1
- 208000002250 Hematologic Neoplasms Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000691589 Homo sapiens 1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase gamma-2 Proteins 0.000 description 1
- 101000623543 Homo sapiens 40S ribosomal protein S15 Proteins 0.000 description 1
- 101000690301 Homo sapiens Aldo-keto reductase family 1 member C4 Proteins 0.000 description 1
- 101001060231 Homo sapiens F-box/WD repeat-containing protein 7 Proteins 0.000 description 1
- 101000935587 Homo sapiens Flavin reductase (NADPH) Proteins 0.000 description 1
- 101001033233 Homo sapiens Interleukin-10 Proteins 0.000 description 1
- 101000998146 Homo sapiens Interleukin-17A Proteins 0.000 description 1
- 101000741885 Homo sapiens Protection of telomeres protein 1 Proteins 0.000 description 1
- 101001116548 Homo sapiens Protein CBFA2T1 Proteins 0.000 description 1
- 101000857677 Homo sapiens Runt-related transcription factor 1 Proteins 0.000 description 1
- 101000984753 Homo sapiens Serine/threonine-protein kinase B-raf Proteins 0.000 description 1
- 101000707567 Homo sapiens Splicing factor 3B subunit 1 Proteins 0.000 description 1
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 1
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 1
- 238000012450 HuMAb Mouse Methods 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 1
- VSNHCAURESNICA-UHFFFAOYSA-N Hydroxyurea Chemical compound NC(=O)NO VSNHCAURESNICA-UHFFFAOYSA-N 0.000 description 1
- MPBVHIBUJCELCL-UHFFFAOYSA-N Ibandronate Chemical compound CCCCCN(C)CCC(O)(P(O)(O)=O)P(O)(O)=O MPBVHIBUJCELCL-UHFFFAOYSA-N 0.000 description 1
- 108700005091 Immunoglobulin Genes Proteins 0.000 description 1
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 108090000176 Interleukin-13 Proteins 0.000 description 1
- 102000003816 Interleukin-13 Human genes 0.000 description 1
- 102100033461 Interleukin-17A Human genes 0.000 description 1
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 1
- 241000235649 Kluyveromyces Species 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- 229930182816 L-glutamine Natural products 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- 239000005511 L01XE05 - Sorafenib Substances 0.000 description 1
- 239000002067 L01XE06 - Dasatinib Substances 0.000 description 1
- 239000005536 L01XE08 - Nilotinib Substances 0.000 description 1
- 239000003798 L01XE11 - Pazopanib Substances 0.000 description 1
- 239000002145 L01XE14 - Bosutinib Substances 0.000 description 1
- UIARLYUEJFELEN-LROUJFHJSA-N LSM-1231 Chemical compound C12=C3N4C5=CC=CC=C5C3=C3C(=O)NCC3=C2C2=CC=CC=C2N1[C@]1(C)[C@](CO)(O)C[C@H]4O1 UIARLYUEJFELEN-LROUJFHJSA-N 0.000 description 1
- JLERVPBPJHKRBJ-UHFFFAOYSA-N LY 117018 Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCC3)=CC=2)C2=CC=C(O)C=C2S1 JLERVPBPJHKRBJ-UHFFFAOYSA-N 0.000 description 1
- IVRXNBXKWIJUQB-UHFFFAOYSA-N LY-2157299 Chemical compound CC1=CC=CC(C=2C(=C3CCCN3N=2)C=2C3=CC(=CC=C3N=CC=2)C(N)=O)=N1 IVRXNBXKWIJUQB-UHFFFAOYSA-N 0.000 description 1
- 229920001491 Lentinan Polymers 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- 108010000817 Leuprolide Proteins 0.000 description 1
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 1
- 229940124640 MK-2206 Drugs 0.000 description 1
- 101150053046 MYD88 gene Proteins 0.000 description 1
- VJRAUFKOOPNFIQ-UHFFFAOYSA-N Marcellomycin Natural products C12=C(O)C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C=C2C(C(=O)OC)C(CC)(O)CC1OC(OC1C)CC(N(C)C)C1OC(OC1C)CC(O)C1OC1CC(O)C(O)C(C)O1 VJRAUFKOOPNFIQ-UHFFFAOYSA-N 0.000 description 1
- 229930126263 Maytansine Natural products 0.000 description 1
- 241000219823 Medicago Species 0.000 description 1
- 229920000877 Melamine resin Polymers 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- IVDYZAAPOLNZKG-KWHRADDSSA-N Mepitiostane Chemical compound O([C@@H]1[C@]2(CC[C@@H]3[C@@]4(C)C[C@H]5S[C@H]5C[C@@H]4CC[C@H]3[C@@H]2CC1)C)C1(OC)CCCC1 IVDYZAAPOLNZKG-KWHRADDSSA-N 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 229930192392 Mitomycin Natural products 0.000 description 1
- 241000713869 Moloney murine leukemia virus Species 0.000 description 1
- 241000204795 Muraena helena Species 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 1
- 102100024134 Myeloid differentiation primary response protein MyD88 Human genes 0.000 description 1
- OUSFTKFNBAZUKL-UHFFFAOYSA-N N-(5-{[(5-tert-butyl-1,3-oxazol-2-yl)methyl]sulfanyl}-1,3-thiazol-2-yl)piperidine-4-carboxamide Chemical compound O1C(C(C)(C)C)=CN=C1CSC(S1)=CN=C1NC(=O)C1CCNCC1 OUSFTKFNBAZUKL-UHFFFAOYSA-N 0.000 description 1
- OVBPIULPVIDEAO-UHFFFAOYSA-N N-Pteroyl-L-glutaminsaeure Natural products C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-UHFFFAOYSA-N 0.000 description 1
- ZDZOTLJHXYCWBA-VCVYQWHSSA-N N-debenzoyl-N-(tert-butoxycarbonyl)-10-deacetyltaxol Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C=CC=CC=4)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)C)C(=O)C1=CC=CC=C1 ZDZOTLJHXYCWBA-VCVYQWHSSA-N 0.000 description 1
- 102000002673 NFATC Transcription Factors Human genes 0.000 description 1
- 108010018525 NFATC Transcription Factors Proteins 0.000 description 1
- 206010061309 Neoplasm progression Diseases 0.000 description 1
- SYNHCENRCUAUNM-UHFFFAOYSA-N Nitrogen mustard N-oxide hydrochloride Chemical compound Cl.ClCC[N+]([O-])(C)CCCl SYNHCENRCUAUNM-UHFFFAOYSA-N 0.000 description 1
- KGTDRFCXGRULNK-UHFFFAOYSA-N Nogalamycin Natural products COC1C(OC)(C)C(OC)C(C)OC1OC1C2=C(O)C(C(=O)C3=C(O)C=C4C5(C)OC(C(C(C5O)N(C)C)O)OC4=C3C3=O)=C3C=C2C(C(=O)OC)C(C)(O)C1 KGTDRFCXGRULNK-UHFFFAOYSA-N 0.000 description 1
- 102000001759 Notch1 Receptor Human genes 0.000 description 1
- 108010029755 Notch1 Receptor Proteins 0.000 description 1
- 239000004677 Nylon Substances 0.000 description 1
- 229930187135 Olivomycin Natural products 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- PBBRWFOVCUAONR-UHFFFAOYSA-N PP2 Chemical compound C12=C(N)N=CN=C2N(C(C)(C)C)N=C1C1=CC=C(Cl)C=C1 PBBRWFOVCUAONR-UHFFFAOYSA-N 0.000 description 1
- 239000002033 PVDF binder Substances 0.000 description 1
- 229930012538 Paclitaxel Natural products 0.000 description 1
- VREZDOWOLGNDPW-ALTGWBOUSA-N Pancratistatin Chemical compound C1=C2[C@H]3[C@@H](O)[C@H](O)[C@@H](O)[C@@H](O)[C@@H]3NC(=O)C2=C(O)C2=C1OCO2 VREZDOWOLGNDPW-ALTGWBOUSA-N 0.000 description 1
- VREZDOWOLGNDPW-MYVCAWNPSA-N Pancratistatin Natural products O=C1N[C@H]2[C@H](O)[C@H](O)[C@H](O)[C@H](O)[C@@H]2c2c1c(O)c1OCOc1c2 VREZDOWOLGNDPW-MYVCAWNPSA-N 0.000 description 1
- 241001631646 Papillomaviridae Species 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 108010057150 Peplomycin Proteins 0.000 description 1
- 108010079855 Peptide Aptamers Proteins 0.000 description 1
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 1
- 102000004861 Phosphoric Diester Hydrolases Human genes 0.000 description 1
- 108090001050 Phosphoric Diester Hydrolases Proteins 0.000 description 1
- 102000004160 Phosphoric Monoester Hydrolases Human genes 0.000 description 1
- 108090000608 Phosphoric Monoester Hydrolases Proteins 0.000 description 1
- KMSKQZKKOZQFFG-HSUXVGOQSA-N Pirarubicin Chemical compound O([C@H]1[C@@H](N)C[C@@H](O[C@H]1C)O[C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1CCCCO1 KMSKQZKKOZQFFG-HSUXVGOQSA-N 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 241001505332 Polyomavirus sp. Species 0.000 description 1
- 239000004793 Polystyrene Substances 0.000 description 1
- HFVNWDWLWUCIHC-GUPDPFMOSA-N Prednimustine Chemical compound O=C([C@@]1(O)CC[C@H]2[C@H]3[C@@H]([C@]4(C=CC(=O)C=C4CC3)C)[C@@H](O)C[C@@]21C)COC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 HFVNWDWLWUCIHC-GUPDPFMOSA-N 0.000 description 1
- 102100038745 Protection of telomeres protein 1 Human genes 0.000 description 1
- 108010066717 Q beta Replicase Proteins 0.000 description 1
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 1
- 108091030071 RNAI Proteins 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- AHHFEZNOXOZZQA-ZEBDFXRSSA-N Ranimustine Chemical compound CO[C@H]1O[C@H](CNC(=O)N(CCCl)N=O)[C@@H](O)[C@H](O)[C@H]1O AHHFEZNOXOZZQA-ZEBDFXRSSA-N 0.000 description 1
- 206010038997 Retroviral infections Diseases 0.000 description 1
- 241000219061 Rheum Species 0.000 description 1
- OWPCHSCAPHNHAV-UHFFFAOYSA-N Rhizoxin Natural products C1C(O)C2(C)OC2C=CC(C)C(OC(=O)C2)CC2CC2OC2C(=O)OC1C(C)C(OC)C(C)=CC=CC(C)=CC1=COC(C)=N1 OWPCHSCAPHNHAV-UHFFFAOYSA-N 0.000 description 1
- 108010073443 Ribi adjuvant Proteins 0.000 description 1
- 108010083644 Ribonucleases Proteins 0.000 description 1
- 102000006382 Ribonucleases Human genes 0.000 description 1
- 102100025373 Runt-related transcription factor 1 Human genes 0.000 description 1
- 241000235070 Saccharomyces Species 0.000 description 1
- 101100379220 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) API2 gene Proteins 0.000 description 1
- 101100407739 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) PET18 gene Proteins 0.000 description 1
- 206010039491 Sarcoma Diseases 0.000 description 1
- 206010070834 Sensitisation Diseases 0.000 description 1
- 102100027103 Serine/threonine-protein kinase B-raf Human genes 0.000 description 1
- XUIMIQQOPSSXEZ-UHFFFAOYSA-N Silicon Chemical compound [Si] XUIMIQQOPSSXEZ-UHFFFAOYSA-N 0.000 description 1
- 229920000519 Sizofiran Polymers 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- 102100031711 Splicing factor 3B subunit 1 Human genes 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 206010042566 Superinfection Diseases 0.000 description 1
- 108010016672 Syk Kinase Proteins 0.000 description 1
- 230000017274 T cell anergy Effects 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- BXFOFFBJRFZBQZ-QYWOHJEZSA-N T-2 toxin Chemical compound C([C@@]12[C@]3(C)[C@H](OC(C)=O)[C@@H](O)[C@H]1O[C@H]1[C@]3(COC(C)=O)C[C@@H](C(=C1)C)OC(=O)CC(C)C)O2 BXFOFFBJRFZBQZ-QYWOHJEZSA-N 0.000 description 1
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 1
- 210000000662 T-lymphocyte subset Anatomy 0.000 description 1
- 108700012411 TNFSF10 Proteins 0.000 description 1
- 108700042805 TRU-015 Proteins 0.000 description 1
- 239000005463 Tandutinib Substances 0.000 description 1
- CGMTUJFWROPELF-UHFFFAOYSA-N Tenuazonic acid Natural products CCC(C)C1NC(=O)C(=C(C)/O)C1=O CGMTUJFWROPELF-UHFFFAOYSA-N 0.000 description 1
- 102100036407 Thioredoxin Human genes 0.000 description 1
- IWEQQRMGNVVKQW-OQKDUQJOSA-N Toremifene citrate Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O.C1=CC(OCCN(C)C)=CC=C1C(\C=1C=CC=CC=1)=C(\CCCl)C1=CC=CC=C1 IWEQQRMGNVVKQW-OQKDUQJOSA-N 0.000 description 1
- 102000046299 Transforming Growth Factor beta1 Human genes 0.000 description 1
- 101800002279 Transforming growth factor beta-1 Proteins 0.000 description 1
- 241000223259 Trichoderma Species 0.000 description 1
- UMILHIMHKXVDGH-UHFFFAOYSA-N Triethylene glycol diglycidyl ether Chemical compound C1OC1COCCOCCOCCOCC1CO1 UMILHIMHKXVDGH-UHFFFAOYSA-N 0.000 description 1
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 1
- 102100040247 Tumor necrosis factor Human genes 0.000 description 1
- 102000014384 Type C Phospholipases Human genes 0.000 description 1
- 108010079194 Type C Phospholipases Proteins 0.000 description 1
- 102100038183 Tyrosine-protein kinase SYK Human genes 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 1
- 108020000999 Viral RNA Proteins 0.000 description 1
- 238000012452 Xenomouse strains Methods 0.000 description 1
- ZYVSOIYQKUDENJ-ASUJBHBQSA-N [(2R,3R,4R,6R)-6-[[(6S,7S)-6-[(2S,4R,5R,6R)-4-[(2R,4R,5R,6R)-4-[(2S,4S,5S,6S)-5-acetyloxy-4-hydroxy-4,6-dimethyloxan-2-yl]oxy-5-hydroxy-6-methyloxan-2-yl]oxy-5-hydroxy-6-methyloxan-2-yl]oxy-7-[(3S,4R)-3,4-dihydroxy-1-methoxy-2-oxopentyl]-4,10-dihydroxy-3-methyl-5-oxo-7,8-dihydro-6H-anthracen-2-yl]oxy]-4-[(2R,4R,5R,6R)-4-hydroxy-5-methoxy-6-methyloxan-2-yl]oxy-2-methyloxan-3-yl] acetate Chemical class COC([C@@H]1Cc2cc3cc(O[C@@H]4C[C@@H](O[C@@H]5C[C@@H](O)[C@@H](OC)[C@@H](C)O5)[C@H](OC(C)=O)[C@@H](C)O4)c(C)c(O)c3c(O)c2C(=O)[C@H]1O[C@H]1C[C@@H](O[C@@H]2C[C@@H](O[C@H]3C[C@](C)(O)[C@@H](OC(C)=O)[C@H](C)O3)[C@H](O)[C@@H](C)O2)[C@H](O)[C@@H](C)O1)C(=O)[C@@H](O)[C@@H](C)O ZYVSOIYQKUDENJ-ASUJBHBQSA-N 0.000 description 1
- UZQJVUCHXGYFLQ-AYDHOLPZSA-N [(2s,3r,4s,5r,6r)-4-[(2s,3r,4s,5r,6r)-4-[(2r,3r,4s,5r,6r)-4-[(2s,3r,4s,5r,6r)-3,5-dihydroxy-6-(hydroxymethyl)-4-[(2s,3r,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxyoxan-2-yl]oxy-3,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-3,5-dihydroxy-6-(hy Chemical compound O([C@H]1[C@H](O)[C@@H](CO)O[C@H]([C@@H]1O)O[C@H]1[C@H](O)[C@@H](CO)O[C@H]([C@@H]1O)O[C@H]1CC[C@]2(C)[C@H]3CC=C4[C@@]([C@@]3(CC[C@H]2[C@@]1(C=O)C)C)(C)CC(O)[C@]1(CCC(CC14)(C)C)C(=O)O[C@H]1[C@@H]([C@@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O[C@H]4[C@@H]([C@@H](O[C@H]5[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O5)O)[C@H](O)[C@@H](CO)O4)O)[C@H](O)[C@@H](CO)O3)O)[C@H](O)[C@@H](CO)O2)O)[C@H](O)[C@@H](CO)O1)O)[C@@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1O UZQJVUCHXGYFLQ-AYDHOLPZSA-N 0.000 description 1
- LUJZZYWHBDHDQX-QFIPXVFZSA-N [(3s)-morpholin-3-yl]methyl n-[4-[[1-[(3-fluorophenyl)methyl]indazol-5-yl]amino]-5-methylpyrrolo[2,1-f][1,2,4]triazin-6-yl]carbamate Chemical compound C=1N2N=CN=C(NC=3C=C4C=NN(CC=5C=C(F)C=CC=5)C4=CC=3)C2=C(C)C=1NC(=O)OC[C@@H]1COCCN1 LUJZZYWHBDHDQX-QFIPXVFZSA-N 0.000 description 1
- SPJCRMJCFSJKDE-ZWBUGVOYSA-N [(3s,8s,9s,10r,13r,14s,17r)-10,13-dimethyl-17-[(2r)-6-methylheptan-2-yl]-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1h-cyclopenta[a]phenanthren-3-yl] 2-[4-[bis(2-chloroethyl)amino]phenyl]acetate Chemical compound O([C@@H]1CC2=CC[C@H]3[C@@H]4CC[C@@H]([C@]4(CC[C@@H]3[C@@]2(C)CC1)C)[C@H](C)CCCC(C)C)C(=O)CC1=CC=C(N(CCCl)CCCl)C=C1 SPJCRMJCFSJKDE-ZWBUGVOYSA-N 0.000 description 1
- IFJUINDAXYAPTO-UUBSBJJBSA-N [(8r,9s,13s,14s,17s)-17-[2-[4-[4-[bis(2-chloroethyl)amino]phenyl]butanoyloxy]acetyl]oxy-13-methyl-6,7,8,9,11,12,14,15,16,17-decahydrocyclopenta[a]phenanthren-3-yl] benzoate Chemical compound C([C@@H]1[C@@H](C2=CC=3)CC[C@]4([C@H]1CC[C@@H]4OC(=O)COC(=O)CCCC=1C=CC(=CC=1)N(CCCl)CCCl)C)CC2=CC=3OC(=O)C1=CC=CC=C1 IFJUINDAXYAPTO-UUBSBJJBSA-N 0.000 description 1
- IHGLINDYFMDHJG-UHFFFAOYSA-N [2-(4-methoxyphenyl)-3,4-dihydronaphthalen-1-yl]-[4-(2-pyrrolidin-1-ylethoxy)phenyl]methanone Chemical compound C1=CC(OC)=CC=C1C(CCC1=CC=CC=C11)=C1C(=O)C(C=C1)=CC=C1OCCN1CCCC1 IHGLINDYFMDHJG-UHFFFAOYSA-N 0.000 description 1
- XZSRRNFBEIOBDA-CFNBKWCHSA-N [2-[(2s,4s)-4-[(2r,4s,5s,6s)-4-amino-5-hydroxy-6-methyloxan-2-yl]oxy-2,5,12-trihydroxy-7-methoxy-6,11-dioxo-3,4-dihydro-1h-tetracen-2-yl]-2-oxoethyl] 2,2-diethoxyacetate Chemical compound O([C@H]1C[C@](CC2=C(O)C=3C(=O)C4=CC=CC(OC)=C4C(=O)C=3C(O)=C21)(O)C(=O)COC(=O)C(OCC)OCC)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 XZSRRNFBEIOBDA-CFNBKWCHSA-N 0.000 description 1
- UOFYSRZSLXWIQB-UHFFFAOYSA-N abivertinib Chemical compound C1CN(C)CCN1C(C(=C1)F)=CC=C1NC1=NC(OC=2C=C(NC(=O)C=C)C=CC=2)=C(C=CN2)C2=N1 UOFYSRZSLXWIQB-UHFFFAOYSA-N 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- ZOZKYEHVNDEUCO-XUTVFYLZSA-N aceglatone Chemical compound O1C(=O)[C@H](OC(C)=O)[C@@H]2OC(=O)[C@@H](OC(=O)C)[C@@H]21 ZOZKYEHVNDEUCO-XUTVFYLZSA-N 0.000 description 1
- 229950002684 aceglatone Drugs 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 229930183665 actinomycin Natural products 0.000 description 1
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 229950004955 adozelesin Drugs 0.000 description 1
- BYRVKDUQDLJUBX-JJCDCTGGSA-N adozelesin Chemical compound C1=CC=C2OC(C(=O)NC=3C=C4C=C(NC4=CC=3)C(=O)N3C[C@H]4C[C@]44C5=C(C(C=C43)=O)NC=C5C)=CC2=C1 BYRVKDUQDLJUBX-JJCDCTGGSA-N 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 108700025316 aldesleukin Proteins 0.000 description 1
- 229960000548 alemtuzumab Drugs 0.000 description 1
- 229940045714 alkyl sulfonate alkylating agent Drugs 0.000 description 1
- 150000008052 alkyl sulfonates Chemical class 0.000 description 1
- SHGAZHPCJJPHSC-YCNIQYBTSA-N all-trans-retinoic acid Chemical compound OC(=O)\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-YCNIQYBTSA-N 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- 239000000956 alloy Substances 0.000 description 1
- 229910045601 alloy Inorganic materials 0.000 description 1
- 229960000473 altretamine Drugs 0.000 description 1
- 229940037003 alum Drugs 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 229960003437 aminoglutethimide Drugs 0.000 description 1
- ROBVIMPUHSLWNV-UHFFFAOYSA-N aminoglutethimide Chemical compound C=1C=C(N)C=CC=1C1(CC)CCC(=O)NC1=O ROBVIMPUHSLWNV-UHFFFAOYSA-N 0.000 description 1
- 229960002749 aminolevulinic acid Drugs 0.000 description 1
- 229960003896 aminopterin Drugs 0.000 description 1
- 229960001220 amsacrine Drugs 0.000 description 1
- XCPGHVQEEXUHNC-UHFFFAOYSA-N amsacrine Chemical compound COC1=CC(NS(C)(=O)=O)=CC=C1NC1=C(C=CC=C2)C2=NC2=CC=CC=C12 XCPGHVQEEXUHNC-UHFFFAOYSA-N 0.000 description 1
- 238000000540 analysis of variance Methods 0.000 description 1
- BBDAGFIXKZCXAH-CCXZUQQUSA-N ancitabine Chemical compound N=C1C=CN2[C@@H]3O[C@H](CO)[C@@H](O)[C@@H]3OC2=N1 BBDAGFIXKZCXAH-CCXZUQQUSA-N 0.000 description 1
- 229950000242 ancitabine Drugs 0.000 description 1
- 239000003098 androgen Substances 0.000 description 1
- 238000009167 androgen deprivation therapy Methods 0.000 description 1
- 229940030486 androgens Drugs 0.000 description 1
- 230000002280 anti-androgenic effect Effects 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 229940124650 anti-cancer therapies Drugs 0.000 description 1
- 229940046836 anti-estrogen Drugs 0.000 description 1
- 230000001833 anti-estrogenic effect Effects 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 230000005809 anti-tumor immunity Effects 0.000 description 1
- 239000000051 antiandrogen Substances 0.000 description 1
- 229940030495 antiandrogen sex hormone and modulator of the genital system Drugs 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 230000007503 antigenic stimulation Effects 0.000 description 1
- 239000013059 antihormonal agent Substances 0.000 description 1
- 229940045687 antimetabolites folic acid analogs Drugs 0.000 description 1
- 239000003816 antisense DNA Substances 0.000 description 1
- OVDSPTSBIQCAIN-UHFFFAOYSA-N ap26113 Chemical compound COC1=CC(N2CCC(CC2)N(C)C)=CC=C1NC(N=1)=NC=C(Cl)C=1NC1=CC=CC=C1P(C)(C)=O OVDSPTSBIQCAIN-UHFFFAOYSA-N 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 239000012736 aqueous medium Substances 0.000 description 1
- 150000008209 arabinosides Chemical class 0.000 description 1
- 206010003246 arthritis Diseases 0.000 description 1
- 229950000847 ascrinvacumab Drugs 0.000 description 1
- 238000002820 assay format Methods 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 230000005784 autoimmunity Effects 0.000 description 1
- 229940120638 avastin Drugs 0.000 description 1
- 229960003005 axitinib Drugs 0.000 description 1
- RITAVMQDGBJQJZ-FMIVXFBMSA-N axitinib Chemical compound CNC(=O)C1=CC=CC=C1SC1=CC=C(C(\C=C\C=2N=CC=CC=2)=NN2)C2=C1 RITAVMQDGBJQJZ-FMIVXFBMSA-N 0.000 description 1
- 229960002756 azacitidine Drugs 0.000 description 1
- VSRXQHXAPYXROS-UHFFFAOYSA-N azanide;cyclobutane-1,1-dicarboxylic acid;platinum(2+) Chemical compound [NH2-].[NH2-].[Pt+2].OC(=O)C1(C(O)=O)CCC1 VSRXQHXAPYXROS-UHFFFAOYSA-N 0.000 description 1
- 229950011321 azaserine Drugs 0.000 description 1
- 150000001541 aziridines Chemical class 0.000 description 1
- 239000002585 base Substances 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 229960003270 belimumab Drugs 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 229960000397 bevacizumab Drugs 0.000 description 1
- 229960000997 bicalutamide Drugs 0.000 description 1
- 229920002988 biodegradable polymer Polymers 0.000 description 1
- 239000004621 biodegradable polymer Substances 0.000 description 1
- 230000008512 biological response Effects 0.000 description 1
- 229950008548 bisantrene Drugs 0.000 description 1
- 229950006844 bizelesin Drugs 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical class N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 229960003736 bosutinib Drugs 0.000 description 1
- UBPYILGKFZZVDX-UHFFFAOYSA-N bosutinib Chemical compound C1=C(Cl)C(OC)=CC(NC=2C3=CC(OC)=C(OCCCN4CCN(C)CC4)C=C3N=CC=2C#N)=C1Cl UBPYILGKFZZVDX-UHFFFAOYSA-N 0.000 description 1
- 238000002725 brachytherapy Methods 0.000 description 1
- KQNZDYYTLMIZCT-KQPMLPITSA-N brefeldin A Chemical compound O[C@@H]1\C=C\C(=O)O[C@@H](C)CCC\C=C\[C@@H]2C[C@H](O)C[C@H]21 KQNZDYYTLMIZCT-KQPMLPITSA-N 0.000 description 1
- JUMGSHROWPPKFX-UHFFFAOYSA-N brefeldin-A Natural products CC1CCCC=CC2(C)CC(O)CC2(C)C(O)C=CC(=O)O1 JUMGSHROWPPKFX-UHFFFAOYSA-N 0.000 description 1
- 229950005993 brivanib alaninate Drugs 0.000 description 1
- LTEJRLHKIYCEOX-UHFFFAOYSA-N brivanib alaninate Chemical compound C1=C2NC(C)=CC2=C(F)C(OC2=NC=NN3C=C(C(=C32)C)OCC(C)OC(=O)C(C)N)=C1 LTEJRLHKIYCEOX-UHFFFAOYSA-N 0.000 description 1
- 229960005520 bryostatin Drugs 0.000 description 1
- MJQUEDHRCUIRLF-TVIXENOKSA-N bryostatin 1 Chemical compound C([C@@H]1CC(/[C@@H]([C@@](C(C)(C)/C=C/2)(O)O1)OC(=O)/C=C/C=C/CCC)=C\C(=O)OC)[C@H]([C@@H](C)O)OC(=O)C[C@H](O)C[C@@H](O1)C[C@H](OC(C)=O)C(C)(C)[C@]1(O)C[C@@H]1C\C(=C\C(=O)OC)C[C@H]\2O1 MJQUEDHRCUIRLF-TVIXENOKSA-N 0.000 description 1
- MUIWQCKLQMOUAT-AKUNNTHJSA-N bryostatin 20 Natural products COC(=O)C=C1C[C@@]2(C)C[C@]3(O)O[C@](C)(C[C@@H](O)CC(=O)O[C@](C)(C[C@@]4(C)O[C@](O)(CC5=CC(=O)O[C@]45C)C(C)(C)C=C[C@@](C)(C1)O2)[C@@H](C)O)C[C@H](OC(=O)C(C)(C)C)C3(C)C MUIWQCKLQMOUAT-AKUNNTHJSA-N 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- MBABCNBNDNGODA-LUVUIASKSA-N bullatacin Chemical compound O1[C@@H]([C@@H](O)CCCCCCCCCC)CC[C@@H]1[C@@H]1O[C@@H]([C@H](O)CCCCCCCCCC[C@@H](O)CC=2C(O[C@@H](C)C=2)=O)CC1 MBABCNBNDNGODA-LUVUIASKSA-N 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- 108700002839 cactinomycin Proteins 0.000 description 1
- 229950009908 cactinomycin Drugs 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- IVFYLRMMHVYGJH-PVPPCFLZSA-N calusterone Chemical compound C1C[C@]2(C)[C@](O)(C)CC[C@H]2[C@@H]2[C@@H](C)CC3=CC(=O)CC[C@]3(C)[C@H]21 IVFYLRMMHVYGJH-PVPPCFLZSA-N 0.000 description 1
- 229950009823 calusterone Drugs 0.000 description 1
- 229940112129 campath Drugs 0.000 description 1
- 229940127093 camptothecin Drugs 0.000 description 1
- VSJKWCGYPAHWDS-FQEVSTJZSA-N camptothecin Chemical compound C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-FQEVSTJZSA-N 0.000 description 1
- 230000009702 cancer cell proliferation Effects 0.000 description 1
- 230000005907 cancer growth Effects 0.000 description 1
- 229960004117 capecitabine Drugs 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- 229960002115 carboquone Drugs 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 229960003261 carmofur Drugs 0.000 description 1
- 229960005243 carmustine Drugs 0.000 description 1
- 229950007509 carzelesin Drugs 0.000 description 1
- BBZDXMBRAFTCAA-AREMUKBSSA-N carzelesin Chemical compound C1=2NC=C(C)C=2C([C@H](CCl)CN2C(=O)C=3NC4=CC=C(C=C4C=3)NC(=O)C3=CC4=CC=C(C=C4O3)N(CC)CC)=C2C=C1OC(=O)NC1=CC=CC=C1 BBZDXMBRAFTCAA-AREMUKBSSA-N 0.000 description 1
- 108010047060 carzinophilin Proteins 0.000 description 1
- 239000005018 casein Substances 0.000 description 1
- BECPQYXYKAMYBN-UHFFFAOYSA-N casein, tech. Chemical compound NCCCCC(C(O)=O)N=C(O)C(CC(O)=O)N=C(O)C(CCC(O)=N)N=C(O)C(CC(C)C)N=C(O)C(CCC(O)=O)N=C(O)C(CC(O)=O)N=C(O)C(CCC(O)=O)N=C(O)C(C(C)O)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=N)N=C(O)C(CCC(O)=O)N=C(O)C(CCC(O)=O)N=C(O)C(COP(O)(O)=O)N=C(O)C(CCC(O)=N)N=C(O)C(N)CC1=CC=CC=C1 BECPQYXYKAMYBN-UHFFFAOYSA-N 0.000 description 1
- 235000021240 caseins Nutrition 0.000 description 1
- 229960002412 cediranib Drugs 0.000 description 1
- 230000020411 cell activation Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 230000004709 cell invasion Effects 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- AEULIVPVIDOLIN-UHFFFAOYSA-N cep-11981 Chemical compound C1=C2C3=C4CNC(=O)C4=C4C5=CN(C)N=C5CCC4=C3N(CC(C)C)C2=CC=C1NC1=NC=CC=N1 AEULIVPVIDOLIN-UHFFFAOYSA-N 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- USVCWSAJUAARAL-MEMLXQNLSA-N chembl551064 Chemical compound C1=2C(N)=NC=NC=2N([C@@H]2C[C@H](C2)N2CCC2)C=C1C(C=1)=CC=CC=1OCC1=CC=CC=C1 USVCWSAJUAARAL-MEMLXQNLSA-N 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 238000000546 chi-square test Methods 0.000 description 1
- 229960004926 chlorobutanol Drugs 0.000 description 1
- 229960001480 chlorozotocin Drugs 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- 230000007012 clinical effect Effects 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 229910017052 cobalt Inorganic materials 0.000 description 1
- 239000010941 cobalt Substances 0.000 description 1
- GUTLYIVDDKVIGB-UHFFFAOYSA-N cobalt atom Chemical compound [Co] GUTLYIVDDKVIGB-UHFFFAOYSA-N 0.000 description 1
- 230000024203 complement activation Effects 0.000 description 1
- 239000002131 composite material Substances 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 238000011340 continuous therapy Methods 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 108010089438 cryptophycin 1 Proteins 0.000 description 1
- PSNOPSMXOBPNNV-VVCTWANISA-N cryptophycin 1 Chemical compound C1=C(Cl)C(OC)=CC=C1C[C@@H]1C(=O)NC[C@@H](C)C(=O)O[C@@H](CC(C)C)C(=O)O[C@H]([C@H](C)[C@@H]2[C@H](O2)C=2C=CC=CC=2)C/C=C/C(=O)N1 PSNOPSMXOBPNNV-VVCTWANISA-N 0.000 description 1
- 108010090203 cryptophycin 8 Proteins 0.000 description 1
- PSNOPSMXOBPNNV-UHFFFAOYSA-N cryptophycin-327 Natural products C1=C(Cl)C(OC)=CC=C1CC1C(=O)NCC(C)C(=O)OC(CC(C)C)C(=O)OC(C(C)C2C(O2)C=2C=CC=CC=2)CC=CC(=O)N1 PSNOPSMXOBPNNV-UHFFFAOYSA-N 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 230000001186 cumulative effect Effects 0.000 description 1
- WZHCOOQXZCIUNC-UHFFFAOYSA-N cyclandelate Chemical compound C1C(C)(C)CC(C)CC1OC(=O)C(O)C1=CC=CC=C1 WZHCOOQXZCIUNC-UHFFFAOYSA-N 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- 229960000684 cytarabine Drugs 0.000 description 1
- 230000002559 cytogenic effect Effects 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- 238000004163 cytometry Methods 0.000 description 1
- 230000000120 cytopathologic effect Effects 0.000 description 1
- 230000007402 cytotoxic response Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- LVXJQMNHJWSHET-AATRIKPKSA-N dacomitinib Chemical compound C=12C=C(NC(=O)\C=C\CN3CCCCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 LVXJQMNHJWSHET-AATRIKPKSA-N 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- 229950006418 dactolisib Drugs 0.000 description 1
- JOGKUKXHTYWRGZ-UHFFFAOYSA-N dactolisib Chemical compound O=C1N(C)C2=CN=C3C=CC(C=4C=C5C=CC=CC5=NC=4)=CC3=C2N1C1=CC=C(C(C)(C)C#N)C=C1 JOGKUKXHTYWRGZ-UHFFFAOYSA-N 0.000 description 1
- 229960002448 dasatinib Drugs 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 229960005052 demecolcine Drugs 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 210000004443 dendritic cell Anatomy 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 229950003913 detorubicin Drugs 0.000 description 1
- WVYXNIXAMZOZFK-UHFFFAOYSA-N diaziquone Chemical compound O=C1C(NC(=O)OCC)=C(N2CC2)C(=O)C(NC(=O)OCC)=C1N1CC1 WVYXNIXAMZOZFK-UHFFFAOYSA-N 0.000 description 1
- 229950002389 diaziquone Drugs 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- UGMCXQCYOVCMTB-UHFFFAOYSA-K dihydroxy(stearato)aluminium Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[Al](O)O UGMCXQCYOVCMTB-UHFFFAOYSA-K 0.000 description 1
- 238000006471 dimerization reaction Methods 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 1
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 1
- 229910000397 disodium phosphate Inorganic materials 0.000 description 1
- 235000019800 disodium phosphate Nutrition 0.000 description 1
- VSJKWCGYPAHWDS-UHFFFAOYSA-N dl-camptothecin Natural products C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)C5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-UHFFFAOYSA-N 0.000 description 1
- 239000003534 dna topoisomerase inhibitor Substances 0.000 description 1
- AMRJKAQTDDKMCE-UHFFFAOYSA-N dolastatin Chemical compound CC(C)C(N(C)C)C(=O)NC(C(C)C)C(=O)N(C)C(C(C)C)C(OC)CC(=O)N1CCCC1C(OC)C(C)C(=O)NC(C=1SC=CN=1)CC1=CC=CC=C1 AMRJKAQTDDKMCE-UHFFFAOYSA-N 0.000 description 1
- 229930188854 dolastatin Natural products 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 229950005778 dovitinib Drugs 0.000 description 1
- ZWAOHEXOSAUJHY-ZIYNGMLESA-N doxifluridine Chemical compound O[C@@H]1[C@H](O)[C@@H](C)O[C@H]1N1C(=O)NC(=O)C(F)=C1 ZWAOHEXOSAUJHY-ZIYNGMLESA-N 0.000 description 1
- 229950005454 doxifluridine Drugs 0.000 description 1
- 229960004679 doxorubicin Drugs 0.000 description 1
- NOTIQUSPUUHHEH-UXOVVSIBSA-N dromostanolone propionate Chemical compound C([C@@H]1CC2)C(=O)[C@H](C)C[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H](OC(=O)CC)[C@@]2(C)CC1 NOTIQUSPUUHHEH-UXOVVSIBSA-N 0.000 description 1
- 239000006196 drop Substances 0.000 description 1
- 229950004683 drostanolone propionate Drugs 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 241001493065 dsRNA viruses Species 0.000 description 1
- 229960005501 duocarmycin Drugs 0.000 description 1
- VQNATVDKACXKTF-XELLLNAOSA-N duocarmycin Chemical compound COC1=C(OC)C(OC)=C2NC(C(=O)N3C4=CC(=O)C5=C([C@@]64C[C@@H]6C3)C=C(N5)C(=O)OC)=CC2=C1 VQNATVDKACXKTF-XELLLNAOSA-N 0.000 description 1
- 229930184221 duocarmycin Natural products 0.000 description 1
- AFMYMMXSQGUCBK-AKMKHHNQSA-N dynemicin a Chemical compound C1#C\C=C/C#C[C@@H]2NC(C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C(O)=C3)=C3[C@@]34O[C@]32[C@@H](C)C(C(O)=O)=C(OC)[C@H]41 AFMYMMXSQGUCBK-AKMKHHNQSA-N 0.000 description 1
- VLCYCQAOQCDTCN-UHFFFAOYSA-N eflornithine Chemical compound NCCCC(N)(C(F)F)C(O)=O VLCYCQAOQCDTCN-UHFFFAOYSA-N 0.000 description 1
- 229960002759 eflornithine Drugs 0.000 description 1
- XOPYFXBZMVTEJF-PDACKIITSA-N eleutherobin Chemical compound C(/[C@H]1[C@H](C(=CC[C@@H]1C(C)C)C)C[C@@H]([C@@]1(C)O[C@@]2(C=C1)OC)OC(=O)\C=C\C=1N=CN(C)C=1)=C2\CO[C@@H]1OC[C@@H](O)[C@@H](O)[C@@H]1OC(C)=O XOPYFXBZMVTEJF-PDACKIITSA-N 0.000 description 1
- XOPYFXBZMVTEJF-UHFFFAOYSA-N eleutherobin Natural products C1=CC2(OC)OC1(C)C(OC(=O)C=CC=1N=CN(C)C=1)CC(C(=CCC1C(C)C)C)C1C=C2COC1OCC(O)C(O)C1OC(C)=O XOPYFXBZMVTEJF-UHFFFAOYSA-N 0.000 description 1
- 229950000549 elliptinium acetate Drugs 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 229950011487 enocitabine Drugs 0.000 description 1
- 229950002189 enzastaurin Drugs 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 1
- 210000002615 epidermis Anatomy 0.000 description 1
- 229960001904 epirubicin Drugs 0.000 description 1
- 229950002973 epitiostanol Drugs 0.000 description 1
- 229930013356 epothilone Natural products 0.000 description 1
- 150000003883 epothilone derivatives Chemical class 0.000 description 1
- 229950009760 epratuzumab Drugs 0.000 description 1
- 229940082789 erbitux Drugs 0.000 description 1
- ITSGNOIFAJAQHJ-BMFNZSJVSA-N esorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)C[C@H](C)O1 ITSGNOIFAJAQHJ-BMFNZSJVSA-N 0.000 description 1
- 229950002017 esorubicin Drugs 0.000 description 1
- LJQQFQHBKUKHIS-WJHRIEJJSA-N esperamicin Chemical compound O1CC(NC(C)C)C(OC)CC1OC1C(O)C(NOC2OC(C)C(SC)C(O)C2)C(C)OC1OC1C(\C2=C/CSSSC)=C(NC(=O)OC)C(=O)C(OC3OC(C)C(O)C(OC(=O)C=4C(=CC(OC)=C(OC)C=4)NC(=O)C(=C)OC)C3)C2(O)C#C\C=C/C#C1 LJQQFQHBKUKHIS-WJHRIEJJSA-N 0.000 description 1
- 229960001842 estramustine Drugs 0.000 description 1
- FRPJXPJMRWBBIH-RBRWEJTLSA-N estramustine Chemical compound ClCCN(CCCl)C(=O)OC1=CC=C2[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CCC2=C1 FRPJXPJMRWBBIH-RBRWEJTLSA-N 0.000 description 1
- 239000000328 estrogen antagonist Substances 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- QSRLNKCNOLVZIR-KRWDZBQOSA-N ethyl (2s)-2-[[2-[4-[bis(2-chloroethyl)amino]phenyl]acetyl]amino]-4-methylsulfanylbutanoate Chemical compound CCOC(=O)[C@H](CCSC)NC(=O)CC1=CC=C(N(CCCl)CCCl)C=C1 QSRLNKCNOLVZIR-KRWDZBQOSA-N 0.000 description 1
- 229960005237 etoglucid Drugs 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 239000013604 expression vector Substances 0.000 description 1
- 229940043168 fareston Drugs 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 229960000961 floxuridine Drugs 0.000 description 1
- ODKNJVUHOIMIIZ-RRKCRQDMSA-N floxuridine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(F)=C1 ODKNJVUHOIMIIZ-RRKCRQDMSA-N 0.000 description 1
- 229960000390 fludarabine Drugs 0.000 description 1
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 229960002074 flutamide Drugs 0.000 description 1
- MKXKFYHWDHIYRV-UHFFFAOYSA-N flutamide Chemical compound CC(C)C(=O)NC1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 MKXKFYHWDHIYRV-UHFFFAOYSA-N 0.000 description 1
- 229960000304 folic acid Drugs 0.000 description 1
- 235000019152 folic acid Nutrition 0.000 description 1
- 239000011724 folic acid Substances 0.000 description 1
- 150000002224 folic acids Chemical class 0.000 description 1
- 229960004783 fotemustine Drugs 0.000 description 1
- YAKWPXVTIGTRJH-UHFFFAOYSA-N fotemustine Chemical compound CCOP(=O)(OCC)C(C)NC(=O)N(CCCl)N=O YAKWPXVTIGTRJH-UHFFFAOYSA-N 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 229950004003 fresolimumab Drugs 0.000 description 1
- 230000007849 functional defect Effects 0.000 description 1
- 229940044658 gallium nitrate Drugs 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 1
- 102000034356 gene-regulatory proteins Human genes 0.000 description 1
- 108091006104 gene-regulatory proteins Proteins 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 229940080856 gleevec Drugs 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 229930182470 glycoside Natural products 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 239000010931 gold Substances 0.000 description 1
- 229910052737 gold Inorganic materials 0.000 description 1
- 229960002913 goserelin Drugs 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 210000004524 haematopoietic cell Anatomy 0.000 description 1
- 230000002489 hematologic effect Effects 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 229940022353 herceptin Drugs 0.000 description 1
- UUVWYPNAQBNQJQ-UHFFFAOYSA-N hexamethylmelamine Chemical compound CN(C)C1=NC(N(C)C)=NC(N(C)C)=N1 UUVWYPNAQBNQJQ-UHFFFAOYSA-N 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 102000052620 human IL10 Human genes 0.000 description 1
- 102000054751 human RUNX1T1 Human genes 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 235000011167 hydrochloric acid Nutrition 0.000 description 1
- 150000003840 hydrochlorides Chemical class 0.000 description 1
- 150000004679 hydroxides Chemical class 0.000 description 1
- 229960001330 hydroxycarbamide Drugs 0.000 description 1
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 1
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 1
- 229940015872 ibandronate Drugs 0.000 description 1
- 229950005646 imgatuzumab Drugs 0.000 description 1
- 210000003297 immature b lymphocyte Anatomy 0.000 description 1
- 230000008102 immune modulation Effects 0.000 description 1
- 229940124644 immune regulator Drugs 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 230000003308 immunostimulating effect Effects 0.000 description 1
- 239000007943 implant Substances 0.000 description 1
- DBIGHPPNXATHOF-UHFFFAOYSA-N improsulfan Chemical compound CS(=O)(=O)OCCCNCCCOS(C)(=O)=O DBIGHPPNXATHOF-UHFFFAOYSA-N 0.000 description 1
- 229950008097 improsulfan Drugs 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 238000005462 in vivo assay Methods 0.000 description 1
- 230000000415 inactivating effect Effects 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 238000011221 initial treatment Methods 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 238000002743 insertional mutagenesis Methods 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 229940079322 interferon Drugs 0.000 description 1
- 229940076144 interleukin-10 Drugs 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 230000037041 intracellular level Effects 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 229940065638 intron a Drugs 0.000 description 1
- 229960005386 ipilimumab Drugs 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 230000000155 isotopic effect Effects 0.000 description 1
- 229940043355 kinase inhibitor Drugs 0.000 description 1
- 238000003368 label free method Methods 0.000 description 1
- 239000004816 latex Substances 0.000 description 1
- 229920000126 latex Polymers 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 229960000681 leflunomide Drugs 0.000 description 1
- 229940115286 lentinan Drugs 0.000 description 1
- 229950001845 lestaurtinib Drugs 0.000 description 1
- GFIJNRVAKGFPGQ-LIJARHBVSA-N leuprolide Chemical compound CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 GFIJNRVAKGFPGQ-LIJARHBVSA-N 0.000 description 1
- 229960004338 leuprorelin Drugs 0.000 description 1
- CMJCXYNUCSMDBY-ZDUSSCGKSA-N lgx818 Chemical compound COC(=O)N[C@@H](C)CNC1=NC=CC(C=2C(=NN(C=2)C(C)C)C=2C(=C(NS(C)(=O)=O)C=C(Cl)C=2)F)=N1 CMJCXYNUCSMDBY-ZDUSSCGKSA-N 0.000 description 1
- 238000007834 ligase chain reaction Methods 0.000 description 1
- 238000012417 linear regression Methods 0.000 description 1
- MPVGZUGXCQEXTM-UHFFFAOYSA-N linifanib Chemical compound CC1=CC=C(F)C(NC(=O)NC=2C=CC(=CC=2)C=2C=3C(N)=NNC=3C=CC=2)=C1 MPVGZUGXCQEXTM-UHFFFAOYSA-N 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 229960002247 lomustine Drugs 0.000 description 1
- 238000010234 longitudinal analysis Methods 0.000 description 1
- 229960003538 lonidamine Drugs 0.000 description 1
- WDRYRZXSPDWGEB-UHFFFAOYSA-N lonidamine Chemical compound C12=CC=CC=C2C(C(=O)O)=NN1CC1=CC=C(Cl)C=C1Cl WDRYRZXSPDWGEB-UHFFFAOYSA-N 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 239000000696 magnetic material Substances 0.000 description 1
- 238000007885 magnetic separation Methods 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- MQXVYODZCMMZEM-ZYUZMQFOSA-N mannomustine Chemical compound ClCCNC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CNCCCl MQXVYODZCMMZEM-ZYUZMQFOSA-N 0.000 description 1
- 229950008612 mannomustine Drugs 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 229950008001 matuzumab Drugs 0.000 description 1
- WKPWGQKGSOKKOO-RSFHAFMBSA-N maytansine Chemical compound CO[C@@H]([C@@]1(O)C[C@](OC(=O)N1)([C@H]([C@@H]1O[C@@]1(C)[C@@H](OC(=O)[C@H](C)N(C)C(C)=O)CC(=O)N1C)C)[H])\C=C\C=C(C)\CC2=CC(OC)=C(Cl)C1=C2 WKPWGQKGSOKKOO-RSFHAFMBSA-N 0.000 description 1
- 229960004961 mechlorethamine Drugs 0.000 description 1
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 1
- JDSHMPZPIAZGSV-UHFFFAOYSA-N melamine Chemical compound NC1=NC(N)=NC(N)=N1 JDSHMPZPIAZGSV-UHFFFAOYSA-N 0.000 description 1
- 229960001924 melphalan Drugs 0.000 description 1
- SGDBTWWWUNNDEQ-LBPRGKRZSA-N melphalan Chemical compound OC(=O)[C@@H](N)CC1=CC=C(N(CCCl)CCCl)C=C1 SGDBTWWWUNNDEQ-LBPRGKRZSA-N 0.000 description 1
- 210000001806 memory b lymphocyte Anatomy 0.000 description 1
- 229950009246 mepitiostane Drugs 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- VJRAUFKOOPNFIQ-TVEKBUMESA-N methyl (1r,2r,4s)-4-[(2r,4s,5s,6s)-5-[(2s,4s,5s,6s)-5-[(2s,4s,5s,6s)-4,5-dihydroxy-6-methyloxan-2-yl]oxy-4-hydroxy-6-methyloxan-2-yl]oxy-4-(dimethylamino)-6-methyloxan-2-yl]oxy-2-ethyl-2,5,7,10-tetrahydroxy-6,11-dioxo-3,4-dihydro-1h-tetracene-1-carboxylat Chemical compound O([C@H]1[C@@H](O)C[C@@H](O[C@H]1C)O[C@H]1[C@H](C[C@@H](O[C@H]1C)O[C@H]1C[C@]([C@@H](C2=CC=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C(O)=C21)C(=O)OC)(O)CC)N(C)C)[C@H]1C[C@H](O)[C@H](O)[C@H](C)O1 VJRAUFKOOPNFIQ-TVEKBUMESA-N 0.000 description 1
- MMNNTJYFHUDSKL-UHFFFAOYSA-N methyl n-[6-[2-(5-chloro-2-methylphenyl)-1-hydroxy-3-oxoisoindol-1-yl]-1h-benzimidazol-2-yl]carbamate Chemical compound C=1C=C2NC(NC(=O)OC)=NC2=CC=1C(C1=CC=CC=C1C1=O)(O)N1C1=CC(Cl)=CC=C1C MMNNTJYFHUDSKL-UHFFFAOYSA-N 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 229950010895 midostaurin Drugs 0.000 description 1
- BMGQWWVMWDBQGC-IIFHNQTCSA-N midostaurin Chemical compound CN([C@H]1[C@H]([C@]2(C)O[C@@H](N3C4=CC=CC=C4C4=C5C(=O)NCC5=C5C6=CC=CC=C6N2C5=C43)C1)OC)C(=O)C1=CC=CC=C1 BMGQWWVMWDBQGC-IIFHNQTCSA-N 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 229960003539 mitoguazone Drugs 0.000 description 1
- MXWHMTNPTTVWDM-NXOFHUPFSA-N mitoguazone Chemical compound NC(N)=N\N=C(/C)\C=N\N=C(N)N MXWHMTNPTTVWDM-NXOFHUPFSA-N 0.000 description 1
- VFKZTMPDYBFSTM-GUCUJZIJSA-N mitolactol Chemical compound BrC[C@H](O)[C@@H](O)[C@@H](O)[C@H](O)CBr VFKZTMPDYBFSTM-GUCUJZIJSA-N 0.000 description 1
- 229950010913 mitolactol Drugs 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 229960000350 mitotane Drugs 0.000 description 1
- 239000003607 modifier Substances 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 229910000403 monosodium phosphate Inorganic materials 0.000 description 1
- 235000019799 monosodium phosphate Nutrition 0.000 description 1
- FOYWNSCCNCUEPU-UHFFFAOYSA-N mopidamol Chemical compound C12=NC(N(CCO)CCO)=NC=C2N=C(N(CCO)CCO)N=C1N1CCCCC1 FOYWNSCCNCUEPU-UHFFFAOYSA-N 0.000 description 1
- 229950010718 mopidamol Drugs 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- ZTFBIUXIQYRUNT-MDWZMJQESA-N mubritinib Chemical compound C1=CC(C(F)(F)F)=CC=C1\C=C\C1=NC(COC=2C=CC(CCCCN3N=NC=C3)=CC=2)=CO1 ZTFBIUXIQYRUNT-MDWZMJQESA-N 0.000 description 1
- 229960000951 mycophenolic acid Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 210000004985 myeloid-derived suppressor cell Anatomy 0.000 description 1
- WPOXAFXHRJYEIC-UHFFFAOYSA-N n-(2-chloro-5-methoxyphenyl)-6-methoxy-7-[(1-methylpiperidin-4-yl)methoxy]quinazolin-4-amine Chemical compound COC1=CC=C(Cl)C(NC=2C3=CC(OC)=C(OCC4CCN(C)CC4)C=C3N=CN=2)=C1 WPOXAFXHRJYEIC-UHFFFAOYSA-N 0.000 description 1
- NJSMWLQOCQIOPE-OCHFTUDZSA-N n-[(e)-[10-[(e)-(4,5-dihydro-1h-imidazol-2-ylhydrazinylidene)methyl]anthracen-9-yl]methylideneamino]-4,5-dihydro-1h-imidazol-2-amine Chemical compound N1CCN=C1N\N=C\C(C1=CC=CC=C11)=C(C=CC=C2)C2=C1\C=N\NC1=NCCN1 NJSMWLQOCQIOPE-OCHFTUDZSA-N 0.000 description 1
- LBWFXVZLPYTWQI-IPOVEDGCSA-N n-[2-(diethylamino)ethyl]-5-[(z)-(5-fluoro-2-oxo-1h-indol-3-ylidene)methyl]-2,4-dimethyl-1h-pyrrole-3-carboxamide;(2s)-2-hydroxybutanedioic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O.CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C LBWFXVZLPYTWQI-IPOVEDGCSA-N 0.000 description 1
- FDMQDKQUTRLUBU-UHFFFAOYSA-N n-[3-[2-[4-(4-methylpiperazin-1-yl)anilino]thieno[3,2-d]pyrimidin-4-yl]oxyphenyl]prop-2-enamide Chemical compound C1CN(C)CCN1C(C=C1)=CC=C1NC1=NC(OC=2C=C(NC(=O)C=C)C=CC=2)=C(SC=C2)C2=N1 FDMQDKQUTRLUBU-UHFFFAOYSA-N 0.000 description 1
- HUFOZJXAKZVRNJ-UHFFFAOYSA-N n-[3-[[2-[4-(4-acetylpiperazin-1-yl)-2-methoxyanilino]-5-(trifluoromethyl)pyrimidin-4-yl]amino]phenyl]prop-2-enamide Chemical compound COC1=CC(N2CCN(CC2)C(C)=O)=CC=C1NC(N=1)=NC=C(C(F)(F)F)C=1NC1=CC=CC(NC(=O)C=C)=C1 HUFOZJXAKZVRNJ-UHFFFAOYSA-N 0.000 description 1
- 229950009708 naquotinib Drugs 0.000 description 1
- 229940086322 navelbine Drugs 0.000 description 1
- IOMMMLWIABWRKL-WUTDNEBXSA-N nazartinib Chemical compound C1N(C(=O)/C=C/CN(C)C)CCCC[C@H]1N1C2=C(Cl)C=CC=C2N=C1NC(=O)C1=CC=NC(C)=C1 IOMMMLWIABWRKL-WUTDNEBXSA-N 0.000 description 1
- 229960000513 necitumumab Drugs 0.000 description 1
- 229950008835 neratinib Drugs 0.000 description 1
- ZNHPZUKZSNBOSQ-BQYQJAHWSA-N neratinib Chemical compound C=12C=C(NC\C=C\CN(C)C)C(OCC)=CC2=NC=C(C#N)C=1NC(C=C1Cl)=CC=C1OCC1=CC=CC=N1 ZNHPZUKZSNBOSQ-BQYQJAHWSA-N 0.000 description 1
- 229940082926 neumega Drugs 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 238000007481 next generation sequencing Methods 0.000 description 1
- 229910052759 nickel Inorganic materials 0.000 description 1
- HHZIURLSWUIHRB-UHFFFAOYSA-N nilotinib Chemical compound C1=NC(C)=CN1C1=CC(NC(=O)C=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)=CC(C(F)(F)F)=C1 HHZIURLSWUIHRB-UHFFFAOYSA-N 0.000 description 1
- 229960001346 nilotinib Drugs 0.000 description 1
- 229960002653 nilutamide Drugs 0.000 description 1
- XWXYUMMDTVBTOU-UHFFFAOYSA-N nilutamide Chemical compound O=C1C(C)(C)NC(=O)N1C1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 XWXYUMMDTVBTOU-UHFFFAOYSA-N 0.000 description 1
- 229950010203 nimotuzumab Drugs 0.000 description 1
- 229960001420 nimustine Drugs 0.000 description 1
- VFEDRRNHLBGPNN-UHFFFAOYSA-N nimustine Chemical compound CC1=NC=C(CNC(=O)N(CCCl)N=O)C(N)=N1 VFEDRRNHLBGPNN-UHFFFAOYSA-N 0.000 description 1
- YMVWGSQGCWCDGW-UHFFFAOYSA-N nitracrine Chemical compound C1=CC([N+]([O-])=O)=C2C(NCCCN(C)C)=C(C=CC=C3)C3=NC2=C1 YMVWGSQGCWCDGW-UHFFFAOYSA-N 0.000 description 1
- 229950008607 nitracrine Drugs 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- KGTDRFCXGRULNK-JYOBTZKQSA-N nogalamycin Chemical compound CO[C@@H]1[C@@](OC)(C)[C@@H](OC)[C@H](C)O[C@H]1O[C@@H]1C2=C(O)C(C(=O)C3=C(O)C=C4[C@@]5(C)O[C@H]([C@H]([C@@H]([C@H]5O)N(C)C)O)OC4=C3C3=O)=C3C=C2[C@@H](C(=O)OC)[C@@](C)(O)C1 KGTDRFCXGRULNK-JYOBTZKQSA-N 0.000 description 1
- 229950009266 nogalamycin Drugs 0.000 description 1
- 102000037979 non-receptor tyrosine kinases Human genes 0.000 description 1
- 108091008046 non-receptor tyrosine kinases Proteins 0.000 description 1
- 239000012740 non-selective inhibitor Substances 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 229920001778 nylon Polymers 0.000 description 1
- 229950005751 ocrelizumab Drugs 0.000 description 1
- CZDBNBLGZNWKMC-MWQNXGTOSA-N olivomycin Chemical class O([C@@H]1C[C@@H](O[C@H](C)[C@@H]1O)OC=1C=C2C=C3C[C@H]([C@@H](C(=O)C3=C(O)C2=C(O)C=1)O[C@H]1O[C@@H](C)[C@H](O)[C@@H](OC2O[C@@H](C)[C@H](O)[C@@H](O)C2)C1)[C@H](OC)C(=O)[C@@H](O)[C@@H](C)O)[C@H]1C[C@H](O)[C@H](OC)[C@H](C)O1 CZDBNBLGZNWKMC-MWQNXGTOSA-N 0.000 description 1
- 229950011093 onapristone Drugs 0.000 description 1
- 230000000771 oncological effect Effects 0.000 description 1
- 108010046821 oprelvekin Proteins 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 150000007530 organic bases Chemical class 0.000 description 1
- 229960003278 osimertinib Drugs 0.000 description 1
- DUYJMQONPNNFPI-UHFFFAOYSA-N osimertinib Chemical compound COC1=CC(N(C)CCN(C)C)=C(NC(=O)C=C)C=C1NC1=NC=CC(C=2C3=CC=CC=C3N(C)C=2)=N1 DUYJMQONPNNFPI-UHFFFAOYSA-N 0.000 description 1
- VYNDHICBIRRPFP-UHFFFAOYSA-N pacific blue Chemical compound FC1=C(O)C(F)=C2OC(=O)C(C(=O)O)=CC2=C1 VYNDHICBIRRPFP-UHFFFAOYSA-N 0.000 description 1
- 229960001592 paclitaxel Drugs 0.000 description 1
- 229960004390 palbociclib Drugs 0.000 description 1
- AHJRHEGDXFFMBM-UHFFFAOYSA-N palbociclib Chemical compound N1=C2N(C3CCCC3)C(=O)C(C(=O)C)=C(C)C2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 AHJRHEGDXFFMBM-UHFFFAOYSA-N 0.000 description 1
- VREZDOWOLGNDPW-UHFFFAOYSA-N pancratistatine Natural products C1=C2C3C(O)C(O)C(O)C(O)C3NC(=O)C2=C(O)C2=C1OCO2 VREZDOWOLGNDPW-UHFFFAOYSA-N 0.000 description 1
- 229960001972 panitumumab Drugs 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000007310 pathophysiology Effects 0.000 description 1
- 229960000639 pazopanib Drugs 0.000 description 1
- CUIHSIWYWATEQL-UHFFFAOYSA-N pazopanib Chemical compound C1=CC2=C(C)N(C)N=C2C=C1N(C)C(N=1)=CC=NC=1NC1=CC=C(C)C(S(N)(=O)=O)=C1 CUIHSIWYWATEQL-UHFFFAOYSA-N 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 229960002340 pentostatin Drugs 0.000 description 1
- FPVKHBSQESCIEP-JQCXWYLXSA-N pentostatin Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=CNC[C@H]2O)=C2N=C1 FPVKHBSQESCIEP-JQCXWYLXSA-N 0.000 description 1
- QIMGFXOHTOXMQP-GFAGFCTOSA-N peplomycin Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCCN[C@@H](C)C=1C=CC=CC=1)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1NC=NC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C QIMGFXOHTOXMQP-GFAGFCTOSA-N 0.000 description 1
- 229950003180 peplomycin Drugs 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 210000001322 periplasm Anatomy 0.000 description 1
- 238000009520 phase I clinical trial Methods 0.000 description 1
- 238000009522 phase III clinical trial Methods 0.000 description 1
- 229960003742 phenol Drugs 0.000 description 1
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 1
- 150000004713 phosphodiesters Chemical class 0.000 description 1
- 235000011007 phosphoric acid Nutrition 0.000 description 1
- 150000003016 phosphoric acids Chemical class 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 229960000952 pipobroman Drugs 0.000 description 1
- NJBFOOCLYDNZJN-UHFFFAOYSA-N pipobroman Chemical compound BrCCC(=O)N1CCN(C(=O)CCBr)CC1 NJBFOOCLYDNZJN-UHFFFAOYSA-N 0.000 description 1
- NUKCGLDCWQXYOQ-UHFFFAOYSA-N piposulfan Chemical compound CS(=O)(=O)OCCC(=O)N1CCN(C(=O)CCOS(C)(=O)=O)CC1 NUKCGLDCWQXYOQ-UHFFFAOYSA-N 0.000 description 1
- 229950001100 piposulfan Drugs 0.000 description 1
- 229960001221 pirarubicin Drugs 0.000 description 1
- 229960003073 pirfenidone Drugs 0.000 description 1
- ISWRGOKTTBVCFA-UHFFFAOYSA-N pirfenidone Chemical compound C1=C(C)C=CC(=O)N1C1=CC=CC=C1 ISWRGOKTTBVCFA-UHFFFAOYSA-N 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 229910052697 platinum Inorganic materials 0.000 description 1
- 150000003057 platinum Chemical class 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 238000003752 polymerase chain reaction Methods 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 239000004800 polyvinyl chloride Substances 0.000 description 1
- 229920000915 polyvinyl chloride Polymers 0.000 description 1
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 239000001103 potassium chloride Substances 0.000 description 1
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M potassium chloride Inorganic materials [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 229960004694 prednimustine Drugs 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 230000001566 pro-viral effect Effects 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- CPTBDICYNRMXFX-UHFFFAOYSA-N procarbazine Chemical compound CNNCC1=CC=C(C(=O)NC(C)C)C=C1 CPTBDICYNRMXFX-UHFFFAOYSA-N 0.000 description 1
- 229960000624 procarbazine Drugs 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 229940087463 proleukin Drugs 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 230000002797 proteolythic effect Effects 0.000 description 1
- WOLQREOUPKZMEX-UHFFFAOYSA-N pteroyltriglutamic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)NC(CCC(=O)NC(CCC(=O)NC(CCC(O)=O)C(O)=O)C(O)=O)C(O)=O)C=C1 WOLQREOUPKZMEX-UHFFFAOYSA-N 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 150000003212 purines Chemical class 0.000 description 1
- 229950010131 puromycin Drugs 0.000 description 1
- 150000003230 pyrimidines Chemical class 0.000 description 1
- 238000011363 radioimmunotherapy Methods 0.000 description 1
- 238000011362 radionuclide therapy Methods 0.000 description 1
- 229960002185 ranimustine Drugs 0.000 description 1
- BMKDZUISNHGIBY-UHFFFAOYSA-N razoxane Chemical compound C1C(=O)NC(=O)CN1C(C)CN1CC(=O)NC(=O)C1 BMKDZUISNHGIBY-UHFFFAOYSA-N 0.000 description 1
- 229960000460 razoxane Drugs 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 229940044551 receptor antagonist Drugs 0.000 description 1
- 239000002464 receptor antagonist Substances 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 239000006215 rectal suppository Substances 0.000 description 1
- 229940100618 rectal suppository Drugs 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 229930002330 retinoic acid Natural products 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- OWPCHSCAPHNHAV-LMONGJCWSA-N rhizoxin Chemical compound C/C([C@H](OC)[C@@H](C)[C@@H]1C[C@H](O)[C@]2(C)O[C@@H]2/C=C/[C@@H](C)[C@]2([H])OC(=O)C[C@@](C2)(C[C@@H]2O[C@H]2C(=O)O1)[H])=C\C=C\C(\C)=C\C1=COC(C)=N1 OWPCHSCAPHNHAV-LMONGJCWSA-N 0.000 description 1
- 229920002477 rna polymer Polymers 0.000 description 1
- 229950004892 rodorubicin Drugs 0.000 description 1
- 238000005096 rolling process Methods 0.000 description 1
- MBABCNBNDNGODA-WPZDJQSSSA-N rolliniastatin 1 Natural products O1[C@@H]([C@@H](O)CCCCCCCCCC)CC[C@H]1[C@H]1O[C@@H]([C@H](O)CCCCCCCCCC[C@@H](O)CC=2C(O[C@@H](C)C=2)=O)CC1 MBABCNBNDNGODA-WPZDJQSSSA-N 0.000 description 1
- VHXNKPBCCMUMSW-FQEVSTJZSA-N rubitecan Chemical compound C1=CC([N+]([O-])=O)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VHXNKPBCCMUMSW-FQEVSTJZSA-N 0.000 description 1
- 229950009919 saracatinib Drugs 0.000 description 1
- OUKYUETWWIPKQR-UHFFFAOYSA-N saracatinib Chemical compound C1CN(C)CCN1CCOC1=CC(OC2CCOCC2)=C(C(NC=2C(=CC=C3OCOC3=2)Cl)=NC=N2)C2=C1 OUKYUETWWIPKQR-UHFFFAOYSA-N 0.000 description 1
- 229930182947 sarcodictyin Natural products 0.000 description 1
- 108010038379 sargramostim Proteins 0.000 description 1
- 229960002530 sargramostim Drugs 0.000 description 1
- 238000005204 segregation Methods 0.000 description 1
- BTIHMVBBUGXLCJ-OAHLLOKOSA-N seliciclib Chemical compound C=12N=CN(C(C)C)C2=NC(N[C@@H](CO)CC)=NC=1NCC1=CC=CC=C1 BTIHMVBBUGXLCJ-OAHLLOKOSA-N 0.000 description 1
- 229950000055 seliciclib Drugs 0.000 description 1
- 229950003647 semaxanib Drugs 0.000 description 1
- 230000008313 sensitization Effects 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 239000010703 silicon Substances 0.000 description 1
- 229910052710 silicon Inorganic materials 0.000 description 1
- 229960000714 sipuleucel-t Drugs 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 229950001403 sizofiran Drugs 0.000 description 1
- 150000003384 small molecules Chemical group 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 229960003787 sorafenib Drugs 0.000 description 1
- IVDHYUQIDRJSTI-UHFFFAOYSA-N sorafenib tosylate Chemical compound [H+].CC1=CC=C(S([O-])(=O)=O)C=C1.C1=NC(C(=O)NC)=CC(OC=2C=CC(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 IVDHYUQIDRJSTI-UHFFFAOYSA-N 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 241000894007 species Species 0.000 description 1
- ICXJVZHDZFXYQC-UHFFFAOYSA-N spongistatin 1 Natural products OC1C(O2)(O)CC(O)C(C)C2CCCC=CC(O2)CC(O)CC2(O2)CC(OC)CC2CC(=O)C(C)C(OC(C)=O)C(C)C(=C)CC(O2)CC(C)(O)CC2(O2)CC(OC(C)=O)CC2CC(=O)OC2C(O)C(CC(=C)CC(O)C=CC(Cl)=C)OC1C2C ICXJVZHDZFXYQC-UHFFFAOYSA-N 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 230000001954 sterilising effect Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 229960001052 streptozocin Drugs 0.000 description 1
- ZSJLQEPLLKMAKR-GKHCUFPYSA-N streptozocin Chemical compound O=NN(C)C(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O ZSJLQEPLLKMAKR-GKHCUFPYSA-N 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 230000010741 sumoylation Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 229940034785 sutent Drugs 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 229960001603 tamoxifen Drugs 0.000 description 1
- 238000004885 tandem mass spectrometry Methods 0.000 description 1
- 229950009893 tandutinib Drugs 0.000 description 1
- UXXQOJXBIDBUAC-UHFFFAOYSA-N tandutinib Chemical compound COC1=CC2=C(N3CCN(CC3)C(=O)NC=3C=CC(OC(C)C)=CC=3)N=CN=C2C=C1OCCCN1CCCCC1 UXXQOJXBIDBUAC-UHFFFAOYSA-N 0.000 description 1
- 229940120982 tarceva Drugs 0.000 description 1
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 1
- RCINICONZNJXQF-XAZOAEDWSA-N taxol® Chemical compound O([C@@H]1[C@@]2(CC(C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3(C21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-XAZOAEDWSA-N 0.000 description 1
- 229940063683 taxotere Drugs 0.000 description 1
- 229950004186 telatinib Drugs 0.000 description 1
- NRUKOCRGYNPUPR-QBPJDGROSA-N teniposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@@H](OC[C@H]4O3)C=3SC=CC=3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 NRUKOCRGYNPUPR-QBPJDGROSA-N 0.000 description 1
- 229960001278 teniposide Drugs 0.000 description 1
- 229960005353 testolactone Drugs 0.000 description 1
- BPEWUONYVDABNZ-DZBHQSCQSA-N testolactone Chemical compound O=C1C=C[C@]2(C)[C@H]3CC[C@](C)(OC(=O)CC4)[C@@H]4[C@@H]3CCC2=C1 BPEWUONYVDABNZ-DZBHQSCQSA-N 0.000 description 1
- RTKIYNMVFMVABJ-UHFFFAOYSA-L thimerosal Chemical compound [Na+].CC[Hg]SC1=CC=CC=C1C([O-])=O RTKIYNMVFMVABJ-UHFFFAOYSA-L 0.000 description 1
- 229940033663 thimerosal Drugs 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 1
- 108060008226 thioredoxin Proteins 0.000 description 1
- 229940094937 thioredoxin Drugs 0.000 description 1
- YFTWHEBLORWGNI-UHFFFAOYSA-N tiamiprine Chemical compound CN1C=NC([N+]([O-])=O)=C1SC1=NC(N)=NC2=C1NC=N2 YFTWHEBLORWGNI-UHFFFAOYSA-N 0.000 description 1
- 229950011457 tiamiprine Drugs 0.000 description 1
- 239000003104 tissue culture media Substances 0.000 description 1
- 229940044693 topoisomerase inhibitor Drugs 0.000 description 1
- 229960000303 topotecan Drugs 0.000 description 1
- UCFGDBYHRUNTLO-QHCPKHFHSA-N topotecan Chemical compound C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 UCFGDBYHRUNTLO-QHCPKHFHSA-N 0.000 description 1
- XFCLJVABOIYOMF-QPLCGJKRSA-N toremifene Chemical compound C1=CC(OCCN(C)C)=CC=C1C(\C=1C=CC=CC=1)=C(\CCCl)C1=CC=CC=C1 XFCLJVABOIYOMF-QPLCGJKRSA-N 0.000 description 1
- 229960005026 toremifene Drugs 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 108700012359 toxins Proteins 0.000 description 1
- FNCMIJWGZNHSBF-UHFFFAOYSA-N trabedersen Chemical compound CC1=CN(C2CC(O)C(COP(=O)(S)OC3CC(OC3COP(=O)(S)OC4CC(OC4COP(=O)(S)OC5CC(OC5COP(=O)(S)OC6CC(OC6COP(=O)(S)OC7CC(OC7COP(=O)(S)OC8CC(OC8COP(=O)(S)OC9CC(OC9COP(=O)(S)OC%10CC(OC%10COP(=O)(S)OC%11CC(OC%11COP(=O)(S)OC%12CC(OC%12COP(=O)(S)OC%13CC(OC%13COP(=O)(S)OC%14CC(OC%14COP(=O)(S)OC%15CC(OC%15CO)N%16C=CC(=NC%16=O)N)n%17cnc%18C(=O)NC(=Nc%17%18)N)n%19cnc%20C(=O)NC(=Nc%19%20)N)N%21C=CC(=NC%21=O)N)n%22cnc%23c(N)ncnc%22%23)N%24C=C(C)C(=O)NC%24=O)n%25cnc%26C(=O)NC(=Nc%25%26)N)N%27C=C(C)C(=O)NC%27=O)N%28C=CC(=NC%28=O)N)N%29C=C(C)C(=O)NC%29=O)n%30cnc%31c(N)ncnc%30%31)N%32C=C(C)C(=O)NC%32=O)N%33C=C(C)C(=O)NC%33=O)O2)C(=O)NC1=O.CC%34=CN(C%35CC(OP(=O)(S)OCC%36OC(CC%36OP(=O)(S)OCC%37OC(CC%37OP(=O)(S)OCC%38OC(CC%38O)n%39cnc%40c(N)ncnc%39%40)N%41C=C(C)C(=O)NC%41=O)n%42cnc%43C(=O)NC(=Nc%42%43)N)C(COP(=O)S)O%35)C(=O)NC%34=O FNCMIJWGZNHSBF-UHFFFAOYSA-N 0.000 description 1
- 229950002824 trabedersen Drugs 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 229940099456 transforming growth factor beta 1 Drugs 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 229960000575 trastuzumab Drugs 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 229950001353 tretamine Drugs 0.000 description 1
- IUCJMVBFZDHPDX-UHFFFAOYSA-N tretamine Chemical compound C1CN1C1=NC(N2CC2)=NC(N2CC2)=N1 IUCJMVBFZDHPDX-UHFFFAOYSA-N 0.000 description 1
- 229960001727 tretinoin Drugs 0.000 description 1
- 229960004560 triaziquone Drugs 0.000 description 1
- PXSOHRWMIRDKMP-UHFFFAOYSA-N triaziquone Chemical compound O=C1C(N2CC2)=C(N2CC2)C(=O)C=C1N1CC1 PXSOHRWMIRDKMP-UHFFFAOYSA-N 0.000 description 1
- 229930013292 trichothecene Natural products 0.000 description 1
- 150000003327 trichothecene derivatives Chemical class 0.000 description 1
- 229960001670 trilostane Drugs 0.000 description 1
- KVJXBPDAXMEYOA-CXANFOAXSA-N trilostane Chemical compound OC1=C(C#N)C[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CC[C@@]32O[C@@H]31 KVJXBPDAXMEYOA-CXANFOAXSA-N 0.000 description 1
- NOYPYLRCIDNJJB-UHFFFAOYSA-N trimetrexate Chemical compound COC1=C(OC)C(OC)=CC(NCC=2C(=C3C(N)=NC(N)=NC3=CC=2)C)=C1 NOYPYLRCIDNJJB-UHFFFAOYSA-N 0.000 description 1
- 229960001099 trimetrexate Drugs 0.000 description 1
- 229950000212 trioxifene Drugs 0.000 description 1
- 229960000875 trofosfamide Drugs 0.000 description 1
- UMKFEPPTGMDVMI-UHFFFAOYSA-N trofosfamide Chemical compound ClCCN(CCCl)P1(=O)OCCCN1CCCl UMKFEPPTGMDVMI-UHFFFAOYSA-N 0.000 description 1
- HDZZVAMISRMYHH-LITAXDCLSA-N tubercidin Chemical compound C1=CC=2C(N)=NC=NC=2N1[C@@H]1O[C@@H](CO)[C@H](O)[C@H]1O HDZZVAMISRMYHH-LITAXDCLSA-N 0.000 description 1
- 230000005748 tumor development Effects 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- 238000010396 two-hybrid screening Methods 0.000 description 1
- 229950009811 ubenimex Drugs 0.000 description 1
- 230000034512 ubiquitination Effects 0.000 description 1
- 238000010798 ubiquitination Methods 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 229960001055 uracil mustard Drugs 0.000 description 1
- 238000001291 vacuum drying Methods 0.000 description 1
- LLDWLPRYLVPDTG-UHFFFAOYSA-N vatalanib succinate Chemical compound OC(=O)CCC(O)=O.C1=CC(Cl)=CC=C1NC(C1=CC=CC=C11)=NN=C1CC1=CC=NC=C1 LLDWLPRYLVPDTG-UHFFFAOYSA-N 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 229960003862 vemurafenib Drugs 0.000 description 1
- GPXBXXGIAQBQNI-UHFFFAOYSA-N vemurafenib Chemical compound CCCS(=O)(=O)NC1=CC=C(F)C(C(=O)C=2C3=CC(=CN=C3NC=2)C=2C=CC(Cl)=CC=2)=C1F GPXBXXGIAQBQNI-UHFFFAOYSA-N 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 1
- 229960004528 vincristine Drugs 0.000 description 1
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 1
- OGWKCGZFUXNPDA-UHFFFAOYSA-N vincristine Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(OC(C)=O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-UHFFFAOYSA-N 0.000 description 1
- 229960004355 vindesine Drugs 0.000 description 1
- UGGWPQSBPIFKDZ-KOTLKJBCSA-N vindesine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(N)=O)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1N=C1[C]2C=CC=C1 UGGWPQSBPIFKDZ-KOTLKJBCSA-N 0.000 description 1
- GBABOYUKABKIAF-IELIFDKJSA-N vinorelbine Chemical compound C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC GBABOYUKABKIAF-IELIFDKJSA-N 0.000 description 1
- 229960002066 vinorelbine Drugs 0.000 description 1
- CILBMBUYJCWATM-PYGJLNRPSA-N vinorelbine ditartrate Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O.OC(=O)[C@H](O)[C@@H](O)C(O)=O.C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC CILBMBUYJCWATM-PYGJLNRPSA-N 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 235000012431 wafers Nutrition 0.000 description 1
- 229940053867 xeloda Drugs 0.000 description 1
- 229950008250 zalutumumab Drugs 0.000 description 1
- 229950009268 zinostatin Drugs 0.000 description 1
- 229960000641 zorubicin Drugs 0.000 description 1
- FBTUMDXHSRTGRV-ALTNURHMSA-N zorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(\C)=N\NC(=O)C=1C=CC=CC=1)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 FBTUMDXHSRTGRV-ALTNURHMSA-N 0.000 description 1
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
- G01N33/57484—Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
Definitions
- the present invention relates to methods for stratification and treatment of progressive chronic lymphocytic in a patient in need thereof
- Chronic lymphocytic leukemia is a lymphoid malignancy prevalent in the elderly presenting with a heterogeneous clinical course. Some patients experience a progressive disease with rapid poor outcome while others exhibit an indolent leukemia that does not impact life expectancy (1). The disease results in the clonal expansion of small, mature B lymphocytes, which accumulate in the bone marrow, blood and secondary lymphoid organs. In progressive patients chemo- and immunotherapies are not curative and residual clonal cancer cells re- populate both lymphoid organs and peripheral blood; the antitumor immune surveillance being ineffective (1, 2). The heterogeneity of CLL has been investigated and related to immunophenotypic markers including CD5, CD 19 or CD38 expressions.
- BCR effectors such as Spleen tyrosine kinase, Phospholipase C g2 or NFAT transcription factors are often overexpressed and/or constitutively activated in B-CLL cells and allow the expression of target genes important for cell survival (5, 6).
- zeta chain associated protein commonly expressed in T and NK cells
- ZAP-70 zeta chain associated protein
- the capacity to generate and propagate such activating signals at the cellular level correlates with disease progression.
- defective signal transduction is prevalent in indolent disease and related to an anergic phenotype of the leukemic cells (7, 8).
- the important driving force of BCR initiated signals is especially encountered in lymph nodes, which are important sites for antigen recognition and account for the accumulation of malignant cells in synergy with microenvironmental factors (9, 10).
- the malignancy is also characterized by an imbalance of the immune subpopulations present in peripheral blood and lymphoid organs of CLL patients.
- T cell subsets that account for immune surveillance in tumor development encompassing CD4 + helpers (Th) and regulatory T cells (Tregs) have an altered ratio in these hematological malignancies (1 1 , 12).
- Th helpers
- Tregs regulatory T cells
- Tregs are increased in CLL and correlate with several clinical/biological features of progressive disease
- CD8 + T cells from CLL patients show functional defect in proliferation and cytotoxicity, but preserved cytokine production reflecting T-cell exhaustion (13).
- murine CD5 + Bla and CDld hlgh CD5 + CDl9 hlgh B10 cells are IL-10 producing cells (22, 23).
- Studies unraveling the functional properties of the CD5 + Bla lineage have uncovered regulatory properties leading to bias of the immune cells repertoire amongst which expansion of the Treg population and suppression of Thl and Thl7 differentiation (24).
- Human Breg subsets, identified by their capacity to suppress Thl differentiation and convert CD4 + T cells into Tregs via IL10 production, have also been described (25, 26).
- various IL10 producing subtypes have been reported (26). The latters include CDl9 + CD24 hlgh CD38 hlgh immature B cells, CD 19 + CD24 high CD27 + B10 cells,
- Induction of IL10 in the various subsets seems to require signals from activated CD4 + T cells with CD40L playing a major stimulatory role, while IL21 -dependant signals rather induce Granzyme (GrB) producing regulatory B cells preferentially (29).
- GrB Granzyme
- Additional induced Bregs can also exert a suppressive mechanism via the production of TGF 1 and indoleamine- 2,3 dioxygenase (IDO) (30).
- CLL-B cells Due to the expression of CD5, CLL-B cells have been hypothesized for long as being derived from a human Bl lineage recognizing natural antibodies (31, 32). At present, CLL-B cells are considered as antigen- experienced B cells with an IGHV mutational status reflecting a T-dependent (mutated IGHV, M-IGHV) or T-independent (unmutated IGHV, UM-IGHV) memory phenotype with expression of CD27 in both cases (1). CLL malignant B cells have a clear survival advantage over the other normal B cells (33). Furthermore, diverse triggering events have been shown to induce IL10 in these cells, which share immunosuppressive capacities with B10 cells (21).
- the inventors systematically analyzed the capacity for CLL cells from donors with indolent and progressive disease to produce in response to BCR/CD40 triggering key cytokines involved in immune modulation and in tumor surveillance.
- Cells that produce these three regulatory proteins display a specific signature somehow different from the already described B10 cells.
- these cells share the regulatory properties attributed to Bregs toward CD4 + T cells.
- we give evidences that the combination of the three factors in a polyfunctional score is indicative of progression and might be used for the stratification of the disease.
- the present invention relates to methods for stratification and treatment of progressive chronic lymphocytic in a patient in need thereof.
- CLL Chronic Lymphocytic Leukemia
- the three proteins are produced by subpopulations with markers of activated and memory B cells defining a specific signature. Based on CD5 and CD 19 expression, intraclonal heterogeneity showed differential regulatory factors production relevant to disease evolution. Functional studies proved their regulatory capacities targeting T cell differentiation, proliferation and secretion. IL-10, TGFp i and FOXP3 expressions combined in a polyfunctional score strongly correlated with high-risk factors of progression. This profiling helps to predict progression and pinpoints immune dysfunction in CLL.
- the invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment comprising a step of determining the level of IL-10, TGFp i and FOXP3 in a biological sample obtained from the patient.
- the method of the invention may further comprises a step consisting of comparing the level of IL-10, TGFp i and FOXP3 in the biological sample with a reference value, wherein detecting differential in the level of IL-10, TGFp i and FOXP3 between the biological sample and the reference value is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
- the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of: i) determining the level of IL-10, TGFp i and FOXP3 in a biological sample obtained from the patient, ii) comparing the level determined at step i) with a reference value, wherein detecting differential in the level of IL-10, TGFp i and FOXP3 between the biological sample and the reference value is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
- the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
- an increase of the levels determined at step 1) and ii) between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
- an increase of the polyfunctional score between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- the method of the invention comprises a step consisting of determining the level of IL-10, TGFp i and FOXP3 in the biological sample, followed by an unsupervised hierarchical cluster analysis and an index called polyfunctional score that was built by combining IL10, TGFp i and FOXP3 expressions.
- An increase of the polyfunctional score between two points of the clinical follow up is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment comprising the steps of: i) determining the expression level of IL-10, TGFp 1 and FOXP3 in a biological sample obtained from the patient, ii) applying an unsupervised hierarchical cluster analysis iii) building a polyfunctional score combining IL-10, TGFp i and FOXP3 expression levels determined at step i), an increase of the polyfunctional score value between two points of the clinical follow up is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- a patient denotes a mammal.
- a patient according to the invention refers to any patient (preferably human) afflicted or at risk to be afflicted with chronic lymphocytic leukemia.
- the term“patient” also refers to any patient (preferably human) afflicted or at risk to be afflicted with progressive chronic lymphocytic leukemia
- chronic lymphocytic leukemia or“CLL” has its general meaning in the art and refers to Acute myeloid leukemia such as revised in the World Health Organisation Classification C91.1.
- the term“Chronic lymphocytic leukemia” also refers to a heterogeneous clonal B cell neoplasm ranging from indolent to rapidly progressive clinical course that eludes tumor clearance.
- the term“Chronic lymphocytic leukemia” also refers to a lymphoid malignancy prevalent in the elderly presenting with a heterogeneous clinical course. Some patients experience a progressive disease with rapid poor outcome while others exhibit an indolent leukemia that does not impact life expectancy (1). The disease results in the clonal expansion of small, mature B lymphocytes, which accumulate in the bone marrow, blood and secondary lymphoid organs.
- IL-10 has its general meaning in the art and refers to Interleukin 10.
- TGFp 1 has its general meaning in the art and refers to Transforming Growth Factor Beta 1.
- FOXP3 has its general meaning in the art and refers to Forkhead Box P3.
- biological sample refers to B cells, chronic lymphocytic leukemia B cells, bone marrow sample, lymphoid organ sample, blood sample, serum sample and plasma sample.
- the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of determining the level of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in a biological sample obtained from the patient.
- the method of the invention may further comprises a step consisting of comparing the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 in the biological sample with a reference value, wherein detecting differential in the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 between the biological sample and the reference value is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
- the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
- detecting differential between the level determined at step i) with the predetermined reference value is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
- the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of: i) determining the level of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in a biological sample obtained from the patient at time tl,
- an increase of the levels determined at step 1) and ii) between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
- an increase of the polyfunctional score between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- the method of the invention may further comprises a step consisting of combining the proportion of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in the biological sample to build a polyfunctional score.
- An increase of the polyfunctional score value between two points of the clinical follow up is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
- the cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 relates to cells expressing high level of IL-10, cells expressing high level of TGFp 1 , and cells expressing high level of FOXP3.
- the term“level of cells expressing a biomarker such as IL-10, TGFp 1 and FOXP3” has its general meaning in the art and refers to the quantity of cells expressing the biomarker or the number of cells expressing the biomarker.
- the term“level of cells expressing a biomarker” also refers to the density of cells expressing a biomarker.
- the term“level of cells expressing a biomarker” also refers to the percentage of cells expressing a biomarker.
- the term “level of cells expressing a biomarker” also refers to the level or percentage of B cells expressing a biomarker.
- the term“level of cells expressing a biomarker” also refers to the level or percentage of CLL B cells expressing a biomarker.
- the term“level of B cells expressing a biomarker” refers to the level or percentage of B cells expressing a biomarker compared to total B cells.
- the term“level of CLL B cells expressing a biomarker” refers to the level or percentage of CLL B cells expressing a bio marker compared to total CLL B cells.
- the“polyfunctional score” refers to a combined value of expression and the term“reference value” refers to a threshold value or a cut-off value.
- the setting of a single “polyfunctional score” or“reference value” thus allows discrimination between a progressive chronic lymphocytic leukemia patient and indolent chronic lymphocytic leukemia patient; discrimination between a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatments and a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia or not resistant to treatments; a poor and a good prognosis with respect to the aggressiveness, invasiveness and/or recurrence of chronic lymphocytic leukemia, cancer relapse and/or overall survival (OS) for a patient.
- OS overall survival
- a “threshold value”, “cut-off value” or“polyfunctional score” can be determined experimentally, empirically, or theoretically.
- a threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art.
- the threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative).
- the optimal sensitivity and specificity (and so the threshold value) can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data.
- ROC Receiver Operating Characteristic
- the threshold value is derived from the quantity (or ratio, or score) determined in a biological sample derived from one or more patients having chronic lymphocytic leukemia. Furthermore, retrospective measurement of the quantity (or ratio, or scores) in properly banked historical patient samples may be used in establishing these threshold values.
- Predetermined reference values used for comparison may comprise “cut-off’ or “threshold” values that may be determined as described herein.
- Each reference (“cut-off’) or polyfunctional score value may be predetermined by carrying out a method comprising the steps of
- step e providing, for each sample provided at step a), information relating to the actual clinical outcome for the corresponding chronic lymphocytic leukemia patient (i.e. indolent chronic lymphocytic leukemia or progressive chronic lymphocytic leukemia; the duration of the event-free survival (EFS), metastasis- free survival (MFS) or the overall survival (OS) or both);
- EFS event-free survival
- MFS metastasis- free survival
- OS overall survival
- the level of the biomarkers or the level of cells expressing the biomarkers has been assessed for 100 cancer samples of 100 patients.
- the 100 samples are ranked according to their level of the biomarkers or their level of cells expressing the biomarkers.
- Sample 1 has the best level of the biomarkers or level of cells expressing the biomarkers and sample 100 has the worst level of the biomarkers or level of cells expressing the biomarkers.
- a first grouping provides two subsets: on one side sample Nr 1 and on the other side the 99 other samples.
- the next grouping provides on one side samples 1 and 2 and on the other side the 98 remaining samples etc., until the last grouping: on one side samples 1 to 99 and on the other side sample Nr 100.
- Kaplan Meier curves are prepared for each of the 99 groups of two subsets. Also for each of the 99 groups, the p value between both subsets was calculated.
- the reference value or polyfunctional score is selected such as the discrimination based on the criterion of the minimum p value is the strongest.
- the level of the biomarkers or level of cells expressing the biomarkers corresponding to the boundary between both subsets for which the p value is minimum is considered as the reference value. It should be noted that the reference value is not necessarily the median value of levels of the m biomarkers or levels of cells expressing the biomarkers.
- the reference value (cut-off value) or polyfunctional score may be used in the present method to discriminate progressive chronic lymphocytic leukemia samples and therefore the corresponding patients.
- Kaplan-Meier curves of percentage of survival as a function of time are commonly to measure the fraction of patients living for a certain amount of time after treatment and are well known by the man skilled in the art.
- a score which is a composite of the level of the biomarkers or level of cells expressing the biomarkers may also be determined and compared to a reference value wherein a difference between said score and said reference value is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
- the score may be generated by a computer program.
- the reference value or the polyfunctional score may correspond to the level of the biomarkers or level of cells expressing the biomarkers determined in a sample associated with a patient having indolent chronic lymphocytic leukemia or a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia.
- a higher level of the biomarkers or level of cells expressing the biomarkers than the reference value or a highest level of the bio markers or level of cells expressing the bio markers in the unsupervised hierarchical cluster analysis is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia, or resisting to treatments and a lower or equal level of the biomarkers or level of cells expressing the biomarkers than the reference value is indicative of a patient having indolent chronic lymphocytic leukemia or a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia.
- the reference value or the polyfunctional score may correspond to the level of the bio markers or level of cells expressing the bio markers determined in a sample associated with a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- a higher or equal level of the biomarkers or level of cells expressing the biomarkers than the reference value or a highest level of the bio markers or level of cells expressing the bio markers in the unsupervised hierarchical cluster analysis is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment
- a lower level of the bio markers or level of cells expressing the bio markers than the reference value is indicative of a patient having indolent chronic lymphocytic leukemia or a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia.
- the method of the invention is performed in at least two times tl and t2. In some embodiments, the method of the invention is performed at time tl and at time t2 different from time tl .
- an increase of the level of the biomarkers, level of cells expressing the biomarkers or the polyfunctional score is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment
- a stable or a decrease of the level of the biomarkers, level of cells expressing the biomarkers or the polyfunctional score is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- the method of the invention further comprises determining the expression level of at least one biomarker selected from the group consisting of pro- inflammatory cytokines and/or regulatory cytokines.
- the method of the invention further comprises determining the level of cells (B cells or CLL B cells) expressing at least one bio marker selected from the group consisting of pro -inflammatory cytokines and/or regulatory cytokines.
- pro -inflammatory cytokines has its general meaning in the art and refers to cytokines or soluble factors that are excreted from immune cells, such as specific helper T cells, and that promote inflammation through pro-inflammatory signals such as IL 1 b, IL2, IL4 or IFNy.
- regulatory cytokines has its general meaning in the art and refers to cytokines or soluble factors that secreted by cells involved in the remodeling of the immune response in normal or pathological environments such as IL6, IL8 or TNFa.
- the equilibrium between positive and negative regulatory cytokines or level of cells expressing regulatory cytokines balancing toward a lower level of pro- inflammatory cytokines or level of cells expressing pro -inflammatory cytokines in favour of the negative regulatory cytokines is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
- a higher level of regulatory cytokines or level of cells expressing regulatory cytokines than the reference value and/or a lower level of pro -inflammatory cytokines or level of cells expressing pro -inflammatory cytokines than the reference value is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia; and a lower level of regulatory cytokines or level of cells expressing regulatory cytokines than the reference value and/or a higher level of pro -inflammatory cytokines or level of cells expressing pro -inflammatory cytokines than the reference value is indicative of a patient having indolent chronic lymphocytic leukemia or a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia.
- the level of cells expressing the bio markers is determined by any well-known method in the art. In some embodiments, the level of cells expressing the biomarkers is determined such as described in the example. In some embodiments, the level of cells expressing the biomarkers is determined by flow cytometry. In some embodiments, the level of cells expressing the biomarkers is determined by IHC or immunofluorescence.
- the quantification of the cells expressing the biomarkers is performed by contacting the biological sample with a binding partner (e.g. an antibody) specific for a cell biomarkers of said cells.
- a binding partner e.g. an antibody
- the quantification of the cells expressing the biomarkers is performed by contacting the tissue tumour tissue sample with a binding partner (e.g. an antibody) specific for IL-10, TGFp 1 or FOXP3.
- the level of cells expressing the bio markers is expressed as the percentage of the specific cells per total cells (set at 100%). In some embodiments, the level of cells expressing the biomarkers may also consist of the number of these cells that are counted per one unit of surface area, e.g. as the number of cells that are counted per mm 2 of surface area of tumour tissue sample. In some embodiments, the quantification of the cells expressing the biomarkers is performed by flow cytometry or Fluorescence-activated cell sorting (FACS). In some embodiments, the quantification of the cells expressing the bio markers is performed by flow cytometry such as described in the example.
- FACS Fluorescence-activated cell sorting
- the level of the biomarkers may be assessed by any of a wide variety of well-known methods for detecting expression of a transcribed nucleic acid or translated protein.
- the biomarker expression level is assessed by analyzing the expression of the protein translated from said gene. Said analysis can be assessed using an antibody (e.g., a radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody), an antibody derivative (e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein of a protein/ligand pair (e.g., biotin-streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically to the protein translated from the gene encoding for the biomarker.
- an antibody e.g., a radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody
- an antibody derivative e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein
- Methods for measuring the expression level of a biomarker in a sample may be assessed by any of a wide variety of well-known methods from one of skill in the art for detecting expression of a protein including, but not limited to, direct methods like mass spectrometry- based quantification methods, protein microarray methods, enzyme immunoassay (EIA), radioimmunoassay (RIA), Immunohistochemistry (IHC), Western blot analysis, ELISA, Luminex, ELISPOT and enzyme linked immunoabsorbant assay and undirect methods based on detecting expression of corresponding messenger ribonucleic acids (mRNAs).
- the mRNA expression profile may be determined by any technology known by a man skilled in the art.
- each mRNA expression level may be measured using any technology known by a man skilled in the art, including nucleic microarrays, quantitative Polymerase Chain Reaction (qPCR), next generation sequencing and hybridization with a labelled probe.
- the binding partner may be an antibody that may be polyclonal or monoclonal, preferably monoclonal (e.g., a isotope-label, element-label, radio-labeled, chromophore- labeled, fluorophore-labeled, or enzyme-labeled antibody), an antibody derivative (e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein of a protein/ligand pair (e.g., biotin-streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically to the protein translated from the gene encoding for the biomarker of the invention.
- monoclonal e.g., a isotope-label, element-label, radio-labeled, chromophore- labeled, fluorophore-labeled, or enzyme-labeled antibody
- the binding partner may be an aptamer.
- the binding partners of the invention such as antibodies or aptamers, may be labelled with a detectable molecule or substance, such as an isotope, an element, a fluorescent molecule, a radioactive molecule or any others labels known in the art. Labels are known in the art that generally provide (either directly or indirectly) a signal.
- the term "labelled", with regard to the antibody is intended to encompass direct labelling of the antibody or aptamer by coupling (i.e., physically linking) a detectable substance, such as an isotope, an element, a radioactive agent or a fluorophore (e.g. fluorescein isothiocyanate (FITC) or phycoerythrin (PE) or Indocyanine (Cy5)) to the antibody or aptamer, as well as indirect labelling of the probe or antibody by reactivity with a detectable substance.
- a detectable substance such as an isotope, an element, a radioactive agent or a fluorophore (e.g. fluorescein isothiocyanate (FITC) or phycoerythrin (PE) or Indocyanine (Cy5)
- FITC fluorescein isothiocyanate
- PE phycoerythrin
- Indocyanine Indocyanine
- radioactive molecules include but are not limited to radioactive atom for scintigraphic studies such as 1123, 1124, Inl l l, Rel86, Rel88, specific isotopes include but are not limited to 13C, 15N, 1261, 79Br, 8lBr.
- the afore mentioned assays generally involve the binding of the binding partner (ie. antibody or aptamer) to a solid support.
- Solid supports which can be used in the practice of the invention include substrates such as nitrocellulose (e. g., in membrane or microtiter well form); polyvinylchloride (e. g., sheets or microtiter wells); polystyrene latex (e.g., beads or microtiter plates); polyvinylidene fluoride; diazotized paper; nylon membranes; activated beads, magnetically responsive beads, silicon wafers.
- substrates such as nitrocellulose (e. g., in membrane or microtiter well form); polyvinylchloride (e. g., sheets or microtiter wells); polystyrene latex (e.g., beads or microtiter plates); polyvinylidene fluoride; diazotized paper; nylon membranes; activated beads, magnetically responsive beads, silicon wa
- an ELISA method can be used, wherein the wells of a microtiter plate are coated with a set of antibodies which recognize said biomarker. A sample containing or suspected of containing said bio marker is then added to the coated wells. After a period of incubation sufficient to allow the formation of antibody-antigen complexes, the plate(s) can be washed to remove unbound moieties and a detectably labelled secondary binding molecule added. The secondary binding molecule is allowed to react with any captured sample marker protein, the plate washed and the presence of the secondary binding molecule detected using methods well known in the art such as Singulex, Quanterix, MSD, Bioscale, Cytof.
- an Enzyme-linked immunospot (ELISpot) method may be used.
- the sample is transferred to a plate which has been coated with the desired anti biomarker capture antibodies.
- Revelation is carried out with biotinylated secondary Abs and standard colorimetric or fluorimetric detection methods such as streptavidin-alkaline phosphatase and NBT-BCIP and the spots counted.
- standard colorimetric or fluorimetric detection methods such as streptavidin-alkaline phosphatase and NBT-BCIP and the spots counted.
- the bead may be a cytometric bead for use in flow cytometry.
- Such beads may for example correspond to BDTM Cytometric Beads commercialized by BD Biosciences (San Jose, California).
- cytometric beads may be suitable for preparing a multiplexed bead assay.
- a multiplexed bead assay such as, for example, the BD(TM) Cytometric Bead Array, is a series of spectrally discrete beads that can be used to capture and quantify soluble antigens.
- beads are labelled with one or more spectrally distinct fluorescent dyes, and detection is carried out using a multiplicity of photodetectors, one for each distinct dye to be detected.
- a number of methods of making and using sets of distinguishable beads have been described in the literature. These include beads distinguishable by size, wherein each size bead is coated with a different target-specific antibody (see e.g.
- beads with two or more fluorescent dyes at varying concentrations wherein the beads are identified by the levels of fluorescence dyes (see e.g. European Patent No. 0 126,450), and beads distinguishably labelled with two different dyes, wherein the beads are identified by separately measuring the fluorescence intensity of each of the dyes (see e.g. U.S. patent Nos. 4,499,052 and 4,717,655).
- fluorescence dyes see e.g. European Patent No. 0 126,450
- beads distinguishably labelled with two different dyes wherein the beads are identified by separately measuring the fluorescence intensity of each of the dyes (see e.g. U.S. patent Nos. 4,499,052 and 4,717,655).
- Examples of one-dimensional arrays of singly dyed beads distinguishable by the level of fluorescence intensity include the BD(TM) Cytometric Bead Array (CBA) (BD Biosciences, San Jose, Calif.) and Cyto-Plex(TM) Flow Cytometry microspheres (Duke Scientific, Palo Alto, Calif.).
- An example of a two-dimensional array of beads distinguishable by a combination of fluorescence intensity (five levels) and size (two sizes) is the QuantumPlex(TM) microspheres (Bangs Laboratories, Fisher, Ind.).
- An example of a two- dimensional array of doubly-dyed beads distinguishable by the levels of fluorescence of each of the two dyes is described in Fulton et al.
- the beads may be labelled with any fluorescent compound known in the art such as e.g. FITC (FL1), PE (FL2), fluorophores for use in the blue laser (e.g. PerCP, PE-Cy7, PE-Cy5, FL3 and APC or Cy5, FL4), fluorophores for use in the red, violet or UV laser (e.g. Pacific blue, pacific orange).
- FL1 FITC
- PE PE
- fluorophores for use in the blue laser e.g. PerCP, PE-Cy7, PE-Cy5, FL3 and APC or Cy5, FL4
- fluorophores for use in the red, violet or UV laser e.g. Pacific blue, pacific orange
- bead is a magnetic bead for use in magnetic separation. Magnetic beads are known to those of skill in the art.
- the magnetic bead is preferably made of a magnetic material selected from the group consisting of metals (e.g.
- bead is bead that is dyed and magnetized.
- protein microarray methods may be used. Typically, at least one antibody or aptamer directed against the biomarker is immobilized or grafted to an array(s), a solid or semi- so lid surface(s). A sample containing or suspected of containing the biomarker is then labelled with at least one isotope or one element or one fluorophore or one colorimetric tag that are not naturally contained in the tested sample. After a period of incubation of said sample with the array sufficient to allow the formation of antibody-antigen complexes, the array is then washed and dried. After all, quantifying said biomarker may be achieved using any appropriate microarray scanner like fluorescence scanner, colorimetric scanner, SIMS (secondary ions mass spectrometry) scanner, maldi scanner, electromagnetic scanner or any technique allowing to quantify said labels.
- any appropriate microarray scanner like fluorescence scanner, colorimetric scanner, SIMS (secondary ions mass spectrometry) scanner
- the antibody or aptamer grafted on the array is labelled.
- reverse phase arrays may be used.
- at least one sample is immobilized or grafted to an array(s), a solid or semi- so lid surface(s).
- An antibody or aptamer against the suspected biomarker is then labelled with at least one isotope or one element or one fluorophore or one colorimetric tag that are not naturally contained in the tested sample.
- the array is then washed and dried.
- detecting quantifying and counting by D-SIMS said biomarker containing said isotope or group of isotopes, and a reference natural element, and then calculating the isotopic ratio between the biomarker and the reference natural element may be achieved using any appropriate microarray scanner like fluorescence scanner, colorimetric scanner, SIMS (secondary ions mass spectrometry) scanner, maldi scanner, electromagnetic scanner or any technique allowing to quantify said labels.
- any appropriate microarray scanner like fluorescence scanner, colorimetric scanner, SIMS (secondary ions mass spectrometry) scanner, maldi scanner, electromagnetic scanner or any technique allowing to quantify said labels.
- said direct analysis can also be assessed by mass Spectrometry.
- Mass spectrometry-based quantification methods may be performed using either labelled or unlabelled approaches (DeSouza and Siu, 2012). Mass spectrometry-based quantification methods may be performed using chemical labeling, metabolic labelingor proteolytic labeling. Mass spectrometry-based quantification methods may be performed using mass spectrometry label free quantification, LTQ Orbitrap Velos, LTQ-MS/MS, a quantification based on extracted ion chromatogram EIC (progenesis LC-MS, Liquid chromatography-mass spectrometry) and then profile alignment to determine differential expression of the biomarker.
- the biomarker expression level is assessed by analyzing the expression of mRNA transcript or mRNA precursors, such as nascent RNA, of biomarker gene. Said analysis can be assessed by preparing mRNA/cDNA from cells in a sample from a subject, and hybridizing the mRNA/cDNA with a reference polynucleotide. The prepared mRNA/cDNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses, such as quantitative PCR (TaqMan), and probes arrays such as GeneChip(TM) DNA Arrays ( AFF YMETRIX) .
- mRNA transcript or mRNA precursors such as nascent RNA
- the analysis of the expression level of mRNA transcribed from the gene encoding for biomarkers involves the process of nucleic acid amplification, e. g., by RT- PCR (the experimental embodiment set forth in U. S. Patent No. 4,683, 202), ligase chain reaction (Barany, 1991), self- sustained sequence replication (Guatelli et al., 1990), transcriptional amplification system (Kwoh et al., 1989), Q-Beta Replicase (Lizardi et al., 1988), rolling circle replication (U. S. Patent No. 5,854, 033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art.
- RT- PCR the experimental embodiment set forth in U. S. Patent No. 4,683, 202
- ligase chain reaction Barany, 1991
- self- sustained sequence replication (Guatelli et al., 1990)
- transcriptional amplification system Kw
- amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5' or 3' regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between.
- amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers.
- the method of the present invention is suitable for monitoring chronic lymphocytic leukemia progression in a patient in need thereof.
- the method of the present invention is suitable for determining whether a patient is eligible or not to an anti-cancer treatment or an anti-cancer therapy. For example, when it is determined that the patient is having or at risk of having or developing progressive chronic lymphocytic leukemia then the physician can take the choice to administer the patient with an anti-cancer treatment.
- the treatment includes chemotherapy, radiotherapy, radio immunotherapy, immunotherapy and drugs suitable for the treatment and prevention of progressive chronic lymphocytic leukemia.
- anti-cancer treatment or“anti-cancer therapy” has its general meaning in the art and refers to anti-cancer compounds used in anti-cancer therapy such as tyrosine kinase inhibitors, tyrosine kinase receptor (TKR) inhibitors, EGFR tyrosine kinase inhibitors, anti- EGFR compounds, anti-HER2 compounds, Vascular Endothelial Growth Factor Receptors (VEGFRs) pathway inhibitors, interferon therapy, alkylating agents, anti-metabolites, immunotherapeutic agents such as sipuleucel-T, Androgen deprivation therapy (ADT), Interferons (IFNs), Interleukins, radiotherapeutic agents (such as Ra223, Pb2l2) and chemotherapeutic agents such as described below.
- TTR tyrosine kinase receptor
- EGFR tyrosine kinase inhibitors anti-EGFR compounds
- anti-HER2 compounds anti-HER2 compounds
- tyrosine kinase inhibitor or“TK ⁇ ” has its general meaning in the art and refers to any of a variety of therapeutic agents or drugs such as compounds inhibiting tyrosine kinase, tyrosine kinase receptor inhibitors (TKRI), EGFR tyrosine kinase inhibitors, EGFR antagonists.
- TKRI tyrosine kinase receptor inhibitors
- EGFR tyrosine kinase inhibitors EGFR antagonists.
- tyrosine kinase inhibitor or“TKI” has its general meaning in the art and refers to any of a variety of therapeutic agents or drugs that act as selective or non-selective inhibitors of receptor and/or non-receptor tyrosine kinases.
- Tyrosine kinase inhibitors and related compounds are well known in the art and described in U.S Patent Publication 2007/0254295, which is incorporated by reference herein in its entirety. It will be appreciated by one of skill in the art that a compound related to a tyrosine kinase inhibitor will recapitulate the effect of the tyrosine kinase inhibitor, e.g., the related compound will act on a different member of the tyrosine kinase signaling pathway to produce the same effect as would a tyrosine kinase inhibitor of that tyrosine kinase.
- tyrosine kinase inhibitors and related compounds suitable for use in methods of embodiments of the present invention include, but are not limited to Erlotinib, sunitinib (Sutent; SU11248), dasatinib (BMS-354825), PP2, BEZ235, saracatinib, gefitinib (Iressa), erlotinib (Tarceva; OSI-1774), lapatinib (GW572016; GW2016), canertinib (Cl 1033), semaxinib (SU5416), vatalanib (PTK787/ZK222584), sorafenib (BAY 43-9006), imatinib (Gleevec; STI571), leflunomide (SU101), vandetanib (Zactima; ZD6474), MK-2206 (8-[4-aminocyclobutyl)phenyl]-9-phenyl-l,2,4-tria
- the tyrosine kinase inhibitor is a small molecule kinase inhibitor that has been orally administered and that has been the subject of at least one Phase I clinical trial, more particularly at least one Phase II clinical, even more particularly at least one Phase III clinical trial, and most particularly approved by the FDA for at least one hematological or oncological indication.
- inhibitors include, but are not limited to Erlotinib, Gefitinib, Lapatinib, Canertinib, BMS-599626 (AC-480), Neratinib, KRN-633, CEP-11981, Imatinib, Nilotinib, Dasatinib, AZM-475271, CP-724714, TAK-165, Sunitinib, Vatalanib, CP- 547632, Vandetanib, Bosutinib, Lestaurtinib, Tandutinib, Midostaurin, Enzastaurin, AEE-788, Pazopanib, Axitinib, Motasenib, OSI-930, Cediranib, KRN-951, Dovitinib, Seliciclib, SNS- 032, PD-0332991, MKC-I (Ro-3 l7453; R-440), Sorafenib, ABT-8
- EGFR tyrosine kinase inhibitors as used herein include, but are not limited to compounds selected from the group consisting of but not limited to Erlotinib, lapatinib, Rociletinib (CO- 1686), gefitinib, Dacomitinib (PF-00299804), Afatanib, Brigatinib (AP26113), WJTOG3405, NEJ002, AZD9291, HM61713, EGF816, ASP 8273, AC 0010.
- antibody EGFR inhibitors examples include Cetuximab, panitumumab, matuzumab, zalutumumab, nimotuzumab, necitumumab, Imgatuzumab (GA201, RO5083945), and ABT- 806.
- chemotherapeutic agent has its general meaning in the art and refers to chemical compounds that are effective in inhibiting tumor growth.
- examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolo melamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues
- calicheamicin especially calicheamicin (11 and calicheamicin 211, see, e.g., Agnew Chem Intl. Ed. Engl. 33: 183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolin
- paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.].) and doxetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-l l ; topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
- anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
- anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide,
- Targeted cancer therapies are drugs or other substances that block the growth and spread of cancer by interfering with specific molecules ("molecular targets") that are involved in the growth, progression, and spread of cancer.
- Targeted cancer therapies are sometimes called “molecularly targeted drugs", “molecularly targeted therapies”, “precision medicines”, or similar names.
- the targeted therapy consists of administering the subject with a tyrosine kinase inhibitor as defined above.
- anti-cancer treatment or “anti-cancer therapy” also refers to immunotherapeutic agent.
- immunotherapeutic agent refers to a compound, composition or treatment that indirectly or directly enhances, stimulates or increases the body's immune response against cancer cells and/or that decreases the side effects of other anticancer therapies. Immunotherapy is thus a therapy that directly or indirectly stimulates or enhances the immune system's responses to cancer cells and/or lessens the side effects that may have been caused by other anti-cancer agents. Immunotherapy is also referred to in the art as immunologic therapy, biological therapy biological response modifier therapy and biotherapy.
- immunotherapeutic agents examples include, but are not limited to, cytokines, cancer vaccines, monoclonal antibodies and non-cytokine adjuvants.
- the immunotherapeutic treatment may consist of administering the subject with an amount of immune cells (T cells, NK, cells, dendritic cells, B cells).
- Immunotherapeutic agents can be non-specific, i.e. boost the immune system generally so that the human body becomes more effective in fighting the growth and/or spread of cancer cells, or they can be specific, i.e. targeted to the cancer cells themselves immunotherapy regimens may combine the use of non-specific and specific immunotherapeutic agents.
- Non-specific immunotherapeutic agents are substances that stimulate or indirectly improve the immune system.
- Non-specific immunotherapeutic agents have been used alone as a main therapy for the treatment of cancer, as well as in addition to a main therapy, in which case the non-specific immunotherapeutic agent functions as an adjuvant to enhance the effectiveness of other therapies (e.g. cancer vaccines).
- Non-specific immunotherapeutic agents can also function in this latter context to reduce the side effects of other therapies, for example, bone marrow suppression induced by certain chemotherapeutic agents.
- Non-specific immunotherapeutic agents can act on key immune system cells and cause secondary responses, such as increased production of cytokines and immunoglobulins. Alternatively, the agents can themselves comprise cytokines.
- Non specific immunotherapeutic agents are generally classified as cytokines or non-cytokine adjuvants.
- cytokines have found application in the treatment of cancer either as general non-specific immunotherapies designed to boost the immune system, or as adjuvants provided with other therapies.
- Suitable cytokines include, but are not limited to, interferons, interleukins and colony- stimulating factors.
- Interferons (IFNs) contemplated by the present invention include the common types of IFNs, IFN-alpha (IFN-a), and IFN-beta (IFN-b). IFNs can act directly on cancer cells, for example, by slowing their growth, promoting their development into cells with more normal behaviour and/or increasing their production of antigens thus making the cancer cells easier for the immune system to recognise and destroy.
- IFNs can also act indirectly on cancer cells, for example, by slowing down angiogenesis, boosting the immune system and/or stimulating natural killer (NK) cells, T cells and macrophages.
- Recombinant IFN-alpha is available commercially as Roferon (Roche Pharmaceuticals) and Intron A (Schering Corporation).
- Interleukins contemplated by the present invention include IL-2, IL-4, IL-l l and IL-12. Examples of commercially available recombinant interleukins include Proleukin® (IL-2; Chiron Corporation) and Neumega® (IL- 12; Wyeth Pharmaceuticals). Zymogenetics, Inc.
- Colony-stimulating factors contemplated by the present invention include sargramostim. Treatment with one or more growth factors can help to stimulate the generation of new blood cells in subjects undergoing traditional chemotherapy. Accordingly, treatment with CSLs can be helpful in decreasing the side effects associated with chemotherapy and can allow for higher doses of chemotherapeutic agents to be used.
- immunotherapeutic agents can be active, i.e. stimulate the body's own immune response, or they can be passive, i.e. comprise immune system components that were generated external to the body.
- Passive specific immunotherapy typically involves the use of one or more monoclonal antibodies that are specific for a particular antigen found on the surface of a cancer cell or that are specific for a particular cell growth factor.
- Monoclonal antibodies may be used in the treatment of cancer in a number of ways, for example, to enhance a subject’s immune response to a specific type of cancer, to interfere with the growth of cancer cells by targeting specific cell growth factors, such as those involved in angiogenesis, or by enhancing the delivery of other anticancer agents to cancer cells when linked or conjugated to agents such as chemotherapeutic agents, radioactive particles or toxins.
- Monoclonal antibodies currently used as cancer immunotherapeutic agents that are suitable for inclusion in the combinations of the present invention include, but are not limited to, rituximab (Rituxan®), trastuzumab (Herceptin®), ibritumomab tiuxetan (Zevalin®), tositumomab (Bexxar®), cetuximab (C-225, Erbitux®), bevacizumab (Avastin®), gemtuzumab ozogamicin (Mylotarg®), alemtuzumab (Campath®), and BL22.
- Other examples include anti-CTLA4 antibodies (e.g.
- antibodies include B cell depleting antibodies.
- Typical B cell depleting antibodies include but are not limited to anti-CD20 monoclonal antibodies [e.g.
- the immunotherapeutic treatment may consist of allografting, in particular, allograft with hematopoietic stem cell HSC.
- the immunotherapeutic treatment may also consist in an adoptive immunotherapy as described by Nicholas P. Restifo, Mark E.
- circulating lymphocytes NK cells
- the activated lymphocytes or NK cells are most particularly be the subject’s own cells that were earlier isolated from a blood or tumor sample and activated (or“expanded”) in vitro.
- anti-cancer treatment or“anti-cancer therapy” also refers to BRAF inhibitors such as vemurafenib, dacarbazine, dabrafenib, BMS-908662, LGX818, PLX3603, RAF265, R05185426, GSK2118436 and compounds described in Morris and Kopetz, 2013.
- BRAF inhibitors such as vemurafenib, dacarbazine, dabrafenib, BMS-908662, LGX818, PLX3603, RAF265, R05185426, GSK2118436 and compounds described in Morris and Kopetz, 2013.
- radiotherapeutic agent as used herein, is intended to refer to any radiotherapeutic agent known to one of skill in the art to be effective to treat or ameliorate cancer, without limitation.
- the radiotherapeutic agent can be an agent such as those administered in brachytherapy or radionuclide therapy such as Ra223 or Pb2l2.
- Such methods can optionally further comprise the administration of one or more additional cancer therapies, such as, but not limited to, chemotherapies, and/or another radiotherapy.
- said active compounds may be contained in the same composition or administrated separately.
- the present invention relates to a compound selected from the group consisting of IL-10 inhibitor, TGFp i inhibitor and/or FOXP3 modulator for use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof
- the present invention relates to a compound selected from the group consisting of IL-10 inhibitor, TGFp i inhibitor and/or FOXP3 modulator for use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof wherein the patient was being classified as having or at risk of having or developing progressive chronic lymphocytic leukemia by the method as above described.
- the invention relates to combined preparation of the compounds of the invention for simultaneous, separate or sequential use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof
- 1, 2 or 3 compounds selected from the group consisting of IL-10 inhibitor, TGFp 1 inhibitor and FOXP3 modulator is used according to the invention.
- the present invention relates to an IL-10 inhibitor for use according to the invention.
- the present invention relates to a TGFp i inhibitor for use according to the invention.
- the present invention relates to a FOXP3 modulator for use according to the invention.
- the present invention relates to an IL-10 inhibitor and TGFp 1 inhibitor for use according to the invention.
- the present invention relates to an IL-10 inhibitor and FOXP3 modulator for use according to the invention.
- the present invention relates to a TGFp i inhibitor and FOXP3 modulator for use according to the invention.
- the present invention relates to an IL-10 inhibitor, TGFp 1 inhibitor and FOXP3 modulator for use according to the invention.
- treatment refers to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of subjects at risk of contracting the disease or suspected to have contracted the disease as well as subjects who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
- the treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.
- therapeutic regimen is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy.
- a therapeutic regimen may include an induction regimen and a maintenance regimen.
- the phrase “induction regimen” or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease.
- the general goal of an induction regimen is to provide a high level of drug to a subject during the initial period of a treatment regimen.
- An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both.
- maintenance regimen refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a subject during treatment of an illness, e.g., to keep the subject in remission for long periods of time (months or years).
- a maintenance regimen may employ continuous therapy (e.g., administering a drug at regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
- modulator has its general meaning in the art and refers to a target inhibitor or activator.
- modulator also refers to a compound that increase or decrease the expression of a specific gene.
- inhibitor has its general meaning in the art and refers to a compound that selectively blocks or inactivates the target (IL-10, TGFp i and/or FOXP3).
- the term“inhibitor” also refers to a compound that selectively blocks the binding of the target to its substrate.
- inhibitor also refers to a compound able to prevent the action of the target for example by inhibiting the target controls of downstream effectors such as inhibiting the activation of the target pathway signalling.
- the term“selectively blocks or inactivates” refers to a compound that preferentially binds to and blocks or inactivates the target with a greater affinity and potency, respectively, than its interaction with the other sub-types of the target family. Compounds that block or inactivate the target, but that may also block or inactivate other target sub-types, as partial or full inhibitors, are contemplated.
- the term“inhibitor” also refers to a compound that inhibits the target expression. Typically, an inhibitor is a small organic molecule, a polypeptide, an aptamer, an antibody, an oligonucleotide or a ribozyme.
- Tests and assays for determining whether a compound is a modulator, an inhibitor or an activator are well known by the skilled person in the art such as described in (Llorente et al, 2000; Clark et al, 2013; WO 2011/143280; US2013/0109619; WO2016196178; WO 2015/124715; US 2009/0325868).
- activator has its general meaning in the art and refers to any compound that can directly or indirectly stimulate the signal transduction cascade related to the target (FOXP3).
- activator also refers to a compound that selectively activates the target.
- a FOXP3 activator is a small organic molecule, a peptide, a modified FOXP3 or an activator of FOXP3 expression.
- a gene product can be the direct transcriptional product of a gene (e.g., mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA or any other type of RNA) or a protein produced by translation of a mRNA.
- Gene products also include messenger RNAs, which are modified, by processes such as capping, polyadenylation, methylation, and editing, and proteins modified by, for example, methylation, acetylation, phosphorylation, ubiquitination, SUMOylation, ADP- ribosylation, myristilation, and glycosylation.
- An“inhibitor of expression” refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene.
- An“activator of expression” refers to a natural or synthetic compound that has a biological effect to activate the expression of a gene.
- IL-10 inhibitor has its general meaning in the art and refers to compounds such as B-N10; ABF 13; B-S10; anti-IL-lO monoclonal antibody; IL-10 receptor antagonists; IL-10 receptor expression inhibitors and compounds described in Llorente et al, 2000; Clark et al, 2013; WO2011/143280; US2013/0109619; WO2016196178.
- TGFP inhibitor has its general meaning in the art and refers to compounds such as Galunisertib (LY2157299); TEW-7197; LY3022859; IMC-TRI; Fresolimumab (GC- 1008); PF-03446962; Trabedersen (AP- 12009); Belagenpumatucel-L; Pirfenidone; and compounds described in Herbertz et al, 2015; W02010/089443; US2011/0294734; US2012/0315256; US2007/0014767; US2013/0225655.
- FOXP3 modulator has its general meaning in the art and refers to compounds such as histone/protein deacetylases (HDAC), histone/protein deacetylases inhibitors (HDACi) and polypeptides and compounds described in US2010/0034786; WO2013/050596; Casares et al., 2010; Lozano et al., 2017.
- the compound of the invention is an aptamer.
- Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition. Aptamers are oligonucleotide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
- Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., 1990.
- the random sequence library is obtainable by combinatorial chemical synthesis of DNA.
- each member is a linear oligomer, eventually chemically modified, of a unique sequence.
- Possible modifications, uses and advantages of this class of molecules have been reviewed in Jayasena S.D., 1999.
- Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al., 1996). Then after raising aptamers directed against the target of the invention as above described, the skilled man in the art can easily select those blocking or inactivating the target.
- the compound of the invention is an antibody (the term including“antibody portion”) directed against the target and which is an IL-10 inhibitor, TGFp 1 inhibitor or a FOXP3 modulator.
- the antibody is a monoclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a polyclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a humanized antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a chimeric antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a light chain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a heavy chain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fab portion of the antibody.
- the portion of the antibody comprises a F(ab')2 portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fc portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fv portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a variable domain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises one or more CDR domains of the antibody.
- antibody includes both naturally occurring and non-naturally occurring antibodies. Specifically, “antibody” includes polyclonal and monoclonal antibodies, and monovalent and divalent fragments thereof. Furthermore, “antibody” includes chimeric antibodies, wholly synthetic antibodies, single chain antibodies, and fragments thereof. The antibody may be a human or nonhuman antibody. A nonhuman antibody may be humanized by recombinant methods to reduce its immunogenicity in man.
- Antibodies are prepared according to conventional methodology. Monoclonal antibodies may be generated using the method of Kohler and Milstein (Nature, 256:495, 1975). To prepare monoclonal antibodies useful in the invention, a mouse or other appropriate host animal is immunized at suitable intervals (e.g., twice-weekly, weekly, twice-monthly or monthly) with antigenic forms of the target. The animal may be administered a final "boost" of antigen within one week of sacrifice. It is often desirable to use an immunologic adjuvant during immunization.
- Suitable immunologic adjuvants include Freund's complete adjuvant, Freund's incomplete adjuvant, alum, Ribi adjuvant, Hunter's Titermax, saponin adjuvants such as QS21 or Quil A, or CpG-containing immunostimulatory oligonucleotides.
- Other suitable adjuvants are well-known in the field.
- the animals may be immunized by subcutaneous, intraperitoneal, intramuscular, intravenous, intranasal or other routes. A given animal may be immunized with multiple forms of the antigen by multiple routes.
- the antigen may be provided as synthetic peptides corresponding to antigenic regions of interest in the target.
- lymphocytes are isolated from the spleen, lymph node or other organ of the animal and fused with a suitable myeloma cell line using an agent such as polyethylene glycol to form a hydridoma.
- cells are placed in media permissive for growth of hybridomas but not the fusion partners using standard methods, as described (Coding, Monoclonal Antibodies: Principles and Practice: Production and Application of Monoclonal Antibodies in Cell Biology, Biochemistry and Immunology, 3rd edition, Academic Press, New York, 1996).
- cell supernatants are analyzed for the presence of antibodies of the desired specificity, i.e., that selectively bind the antigen.
- Suitable analytical techniques include ELISA, flow cytometry, immunoprecipitation, and western blotting. Other screening techniques are well-known in the field. Preferred techniques are those that confirm binding of antibodies to conformationally intact, natively folded antigen, such as non-denaturing ELISA, flow cytometry, and immunoprecipitation.
- the Fc' and Fc regions are effectors of the complement cascade but are not involved in antigen binding.
- an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region designated a Fab fragment
- Fab fragments retains one of the antigen binding sites of an intact antibody molecule.
- Fab fragments consist of a covalently bound antibody light chain and a portion of the antibody heavy chain denoted Fd.
- the Fd fragments are the major determinant of antibody specificity (a single Fd fragment may be associated with up to ten different light chains without altering antibody specificity) and Fd fragments retain epitope-binding ability in isolation.
- CDRs complementarity determining regions
- FRs framework regions
- CDR1 through CDRS complementarity determining regions
- non-CDR regions of a mammalian antibody may be replaced with similar regions of conspecific or heterospecific antibodies while retaining the epitopic specificity of the original antibody. This is most clearly manifested in the development and use of "humanized" antibodies in which non-human CDRs are covalently joined to human FR and/or Fc/pFc’ regions to produce a functional antibody.
- compositions and methods that include humanized forms of antibodies.
- humanized describes antibodies wherein some, most or all of the amino acids outside the CDR regions are replaced with corresponding amino acids derived from human immunoglobulin molecules.
- Methods of humanization include, but are not limited to, those described in U.S. Pat. Nos. 4,816,567, 5,225,539, 5,585,089, 5,693,761, 5,693,762 and 5,859,205, which are hereby incorporated by reference.
- the above U.S. Pat. Nos. 5,585,089 and 5,693,761, and WO 90/07861 also propose four possible criteria which may be used in designing the humanized antibodies.
- the first proposal was that for an acceptor, use a framework from a particular human immunoglobulin that is unusually homologous to the donor immunoglobulin to be humanized, or use a consensus framework from many human antibodies.
- the second proposal was that if an amino acid in the framework of the human immunoglobulin is unusual and the donor amino acid at that position is typical for human sequences, then the donor amino acid rather than the acceptor may be selected.
- the third proposal was that in the positions immediately adjacent to the 3 CDRs in the humanized immunoglobulin chain, the donor amino acid rather than the acceptor amino acid may be selected.
- the fourth proposal was to use the donor amino acid reside at the framework positions at which the amino acid is predicted to have a side chain atom within 3 A of the CDRs in a three dimensional model of the antibody and is predicted to be capable of interacting with the CDRs.
- the above methods are merely illustrative of some of the methods that one skilled in the art could employ to make humanized antibodies.
- One of ordinary skill in the art will be familiar with other methods for antibody humanization.
- humanized forms of the antibodies some, most or all of the amino acids outside the CDR regions have been replaced with amino acids from human immunoglobulin molecules but where some, most or all amino acids within one or more CDR regions are unchanged. Small additions, deletions, insertions, substitutions or modifications of amino acids are permissible as long as they would not abrogate the ability of the antibody to bind a given antigen.
- Suitable human immunoglobulin molecules would include IgGl, IgG2, IgG3, IgG4, IgA and IgM molecules.
- a "humanized" antibody retains a similar antigenic specificity as the original antibody.
- the affinity and/or specificity of binding of the antibody may be increased using methods of "directed evolution", as described by Wu et ah, /. Mol. Biol. 294: 151, 1999, the contents of which are incorporated herein by reference.
- Fully human monoclonal antibodies also can be prepared by immunizing mice transgenic for large portions of human immunoglobulin heavy and light chain loci. See, e.g., U.S. Pat. Nos. 5,591,669, 5,598,369, 5,545,806, 5,545,807, 6,150,584, and references cited therein, the contents of which are incorporated herein by reference. These animals have been genetically modified such that there is a functional deletion in the production of endogenous (e.g., murine) antibodies. The animals are further modified to contain all or a portion of the human germ-line immunoglobulin gene locus such that immunization of these animals will result in the production of fully human antibodies to the antigen of interest.
- monoclonal antibodies can be prepared according to standard hybridoma technology. These monoclonal antibodies will have human immunoglobulin amino acid sequences and therefore will not provoke human anti-mouse antibody (KAMA) responses when administered to humans.
- KAMA human anti-mouse antibody
- the present invention also provides for F(ab') 2 Fab, Fv and Fd fragments; chimeric antibodies in which the Fc and/or FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric F(ab')2 fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric Fab fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; and chimeric Fd fragment antibodies in which the FR and/or CDR1 and/or CDR2 regions have been replaced by homologous human or non human sequences.
- the present invention also includes so-called single chain antibodies.
- the various antibody molecules and fragments may derive from any of the commonly known immunoglobulin classes, including but not limited to IgA, secretory IgA, IgE, IgG and IgM.
- IgG subclasses are also well known to those in the art and include but are not limited to human IgGl, IgG2, IgG3 and IgG4.
- the compound of the invention is a Human IgG4.
- the antibody according to the invention is a single domain antibody.
- the term“single domain antibody” (sdAb) or “VHH” refers to the single heavy chain variable domain of antibodies of the type that can be found in Camelid mammals which are naturally devoid of light chains. Such VHH are also called“nanobody®”. According to the invention, sdAb can particularly be llama sdAb.
- the term“VHH” refers to the single heavy chain having 3 complementarity determining regions (CDRs): CDR1, CDR2 and CDR3.
- the term“complementarity determining region” or“CDR” refers to the hypervariable amino acid sequences which define the binding affinity and specificity of the VHH.
- the VHH according to the invention can readily be prepared by an ordinarily skilled artisan using routine experimentation.
- the VHH variants and modified form thereof may be produced under any known technique in the art such as in-vitro maturation.
- VHHs or sdAbs are usually generated by PCR cloning of the V-domain repertoire from blood, lymph node, or spleen cDNA obtained from immunized animals into a phage display vector, such as pHEN2.
- Antigen- specific VHHs are commonly selected by panning phage libraries on immobilized antigen, e.g., antigen coated onto the plastic surface of a test tube, biotinylated antigens immobilized on streptavidin beads, or membrane proteins expressed on the surface of cells.
- immobilized antigen e.g., antigen coated onto the plastic surface of a test tube, biotinylated antigens immobilized on streptavidin beads, or membrane proteins expressed on the surface of cells.
- VHHs often show lower affinities for their antigen than VHHs derived from animals that have received several immunizations.
- VHHs from immune libraries are attributed to the natural selection of variant VHHs during clonal expansion of B-cells in the lymphoid organs of immunized animals.
- the affinity of VHHs from non-immune libraries can often be improved by mimicking this strategy in vitro, i.e., by site directed mutagenesis of the CDR regions and further rounds of panning on immobilized antigen under conditions of increased stringency (higher temperature, high or low salt concentration, high or low pH, and low antigen concentrations).
- VHHs derived from camelid are readily expressed in and purified from the E. coli periplasm at much higher levels than the corresponding domains of conventional antibodies.
- VHHs generally display high solubility and stability and can also be readily produced in yeast, plant, and mammalian cells.
- the“Hamers patents” describe methods and techniques for generating VHH against any desired target (see for example US 5,800,988; US 5,874, 541 and US 6,015,695).
- The“Hamers patents” more particularly describe production of VHHs in bacterial hosts such as E. coli (see for example US 6,765,087) and in lower eukaryotic hosts such as moulds (for example Aspergillus or Trichoderma) or in yeast (for example Saccharomyces, Kluyveromyces, Hansenula or Pichia) (see for example US 6,838,254).
- the invention provides an antibody that competes for binding to the target with the antibody of the invention.
- binding in the context of the binding of an antibody to a predetermined antigen or epitope typically is a binding with an affinity corresponding to a KD of about 10-7 M or less, such as about 10-8 M or less, such as about 10-9 M or less, about 10- 10 M or less, or about 10-11 M or even less when determined by for instance surface plasmon resonance (SPR) technology in a BIAcore 3000 instrument using a soluble form of the antigen as the ligand and the antibody as the analyte.
- SPR surface plasmon resonance
- BIACORE® GE Healthcare, Piscaataway, NJ
- BIACORE® is one of a variety of surface plasmon resonance assay formats that are routinely used to epitope bin panels of monoclonal antibodies.
- an antibody binds to the predetermined antigen with an affinity corresponding to a KD that is at least ten- fold lower, such as at least lOO-fold lower, for instance at least 1, 000-fold lower, such as at least 10,000-fold lower, for instance at least 100,000-fold lower than its KD for binding to a non-specific antigen (e.g., BSA, casein), which is not identical or closely related to the predetermined antigen.
- a non-specific antigen e.g., BSA, casein
- An antibody is said to essentially not bind an antigen or epitope if such binding is either not detectable (using, for example, plasmon resonance (SPR) technology in a BIAcore 3000 instrument using a soluble form of the antigen as the ligand and the antibody as the analyte), or is 100 fold, 500 fold, 1000 fold or more than 1000 fold less than the binding detected by that antibody and an antigen or epitope having a different chemical structure or amino acid sequence.
- SPR plasmon resonance
- Additional antibodies can be identified based on their ability to cross-compete (e.g., to competitively inhibit the binding of, in a statistically significant manner) with other antibodies of the invention in standard antigen binding assays.
- the ability of a test antibody to inhibit the binding of antibodies of the present invention to the target demonstrates that the test antibody can compete with that antibody for binding to the target; such an antibody may, according to non-limiting theory, bind to the same or a related (e.g., a structurally similar or spatially proximal) epitope on the target as the antibody with which it competes.
- another aspect of the invention provides antibodies that bind to the same antigen as, and compete with, the antibodies disclosed herein.
- the antibodies or antigen binding fragments of the invention bind to one or more epitopes of the target.
- the epitopes to which the present antibodies or antigen binding fragments bind are linear epitopes.
- the epitopes to which the present antibodies or antigen binding fragments bind are non-linear, conformational epitopes.
- the compound of the invention is an IL-10 expression inhibitor, TGFp i expression inhibitor or a FOXP3 expression inhibitor.
- An“inhibitor of expression” refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene.
- the target expression inhibitors for use in the present invention may be based on antisense oligonucleotide constructs.
- Anti-sense oligonucleotides including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of the target mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of the target proteins, and thus activity, in a cell.
- antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding the target can be synthesized, e.g., by conventional phosphodiester techniques and administered by e.g., intravenous injection or infusion.
- Small inhibitory RNAs can also function as target expression inhibitors for use in the present invention.
- the target gene expression can be reduced by contacting the subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that the target expression is specifically inhibited (i.e. RNA interference or RNAi).
- dsRNA small double stranded RNA
- RNAi RNA interference
- Methods for selecting an appropriate dsRNA or dsRNA-encoding vector are well known in the art for genes whose sequence is known (e.g. see Tuschl, T. et al. (1999); Elbashir, S. M. et al. (2001); Hannon, GJ. (2002); McManus, MT. et al.
- Ribozymes can also function as target expression inhibitors for use in the present invention.
- Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
- the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
- Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of the target mRNA sequences are thereby useful within the scope of the present invention.
- ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable. The suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
- antisense oligonucleotides and ribozymes useful target inhibitors can be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules can be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences can be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention can be introduced as a means of increasing intracellular stability and half-life.
- Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3’ ends of the molecule, or the use of phosphorothioate or 2’-0-methyl rather than phosphodiesterase linkages within the oligonucleotide backbone.
- Antisense oligonucleotides siRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector.
- a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide siRNA or ribozyme nucleic acid to the cells and preferably cells expressing the target.
- the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
- the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide siRNA or ribozyme nucleic acid sequences.
- Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus.
- retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
- retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
- adenovirus adeno
- Non-cytopathic viral vectors are based on non-cytopathic eukaryotic viruses in which non- essential genes have been replaced with the gene of interest.
- Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA.
- Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle).
- retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
- Standard protocols for producing replication-deficient retroviruses including the steps of incorporation of exogenous genetic material into a plasmid, transfection of a packaging cell lined with plasmid, production of recombinant retroviruses by the packaging cell line, collection of viral particles from tissue culture media, and infection of the target cells with viral particles
- KRIEGLER A Laboratory Manual
- MURRY Method of Recombinant retroviruses by the packaging cell line
- Methods in Molecular Biology vol.7, Humana Press, Inc., Cliffton, N.J., 1991.
- adeno-viruses and adeno-associated viruses are double-stranded DNA viruses that have already been approved for human use in gene therapy.
- the adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions.
- the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
- adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
- the adeno-associated virus can also function in an extrachromosomal fashion.
- Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g., SANBROOK et al, "Molecular Cloning: A Laboratory Manual," Second Edition, Cold Spring Harbor Laboratory Press, 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid.
- Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
- the DNA plasmid can be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally.
- the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
- the inhibitors according to the invention as described above are administered to the patient in a therapeutically effective amount.
- a “therapeutically effective amount” of the inhibitor of the present invention as above described is meant a sufficient amount of the inhibitor for treating cancer at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the inhibitors and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
- the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific inhibitor employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific inhibitor employed; the duration of the treatment; drugs used in combination or coincidential with the specific inhibitor employed; and like factors well known in the medical arts.
- the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
- the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the inhibitor of the present invention for the symptomatic adjustment of the dosage to the patient to be treated.
- a medicament typically contains from about 0.01 mg to about 500 mg of the inhibitor of the present invention, particularly from 1 mg to about 100 mg of the inhibitor of the present invention.
- An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
- the compound according to the invention may be used in a concentration between 0.01 mM and 20 mM, particularly, the compound of the invention may be used in a concentration of 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 20.0 mM.
- the compound of the present invention is administered to the subject in the form of a pharmaceutical composition.
- the compound of the present invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
- “Pharmaceutically” or “pharmaceutically acceptable” refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate.
- a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
- the active principle in the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings.
- Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
- the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
- vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
- These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
- the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
- the form In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
- Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
- the compound of the present invention can be formulated into a composition in a neutral or salt form.
- Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
- inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
- Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine,
- the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
- the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
- the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
- the composition includes isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
- Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
- dispersions are prepared by incorporating the various sterilized agent of the present inventions into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
- sterile powders for the preparation of sterile injectable solutions the typical methods of preparation are vacuum-drying and freeze- drying techniques which yield a powder of the compound of the present invention plus any additional desired ingredient from a previously sterile- filtered solution thereof.
- the preparation of more, or highly concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small tumor area.
- solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
- the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
- aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
- aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
- sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
- the compound of the present invention is administered sequentially or concomitantly with one or more therapeutic active agent such as anti-cancer therapy such as immunotherapeutic agent, chemotherapeutic agent or radiotherapeutic agent.
- therapeutic active agent such as anti-cancer therapy
- immunotherapeutic agent such as immunotherapeutic agent, chemotherapeutic agent or radiotherapeutic agent.
- said additional active compounds may be contained in the same composition or administrated separately.
- the pharmaceutical composition of the invention relates to combined preparation for simultaneous, separate or sequential use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof.
- kits comprising the compound of the invention. Kits containing the compound of the invention find use in therapeutic methods.
- the present invention relates to a method for treating progressive chronic lymphocytic leukemia in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound selected from the group consisting of IL-10 inhibitor, TGFp 1 inhibitor and/or FOXP3 modulator.
- the present invention relates to a method of treating progressive chronic lymphocytic leukemia in a patient in need thereof comprising the steps of:
- the present invention relates to a method of screening a candidate compound for use as a drug for treating cancer in a patient in need thereof, wherein the method comprises the steps of:
- IL-10 providing a IL-10, TGFp i and/or FOXP3, providing a cell, B cell, chronic lymphocytic leukemia B cells, bone marrow sample, lymphoid organ sample, blood sample, serum sample and plasma sample, tissue sample or organism expressing IL-10, TGFp i and/or FOXP3, providing a candidate compound such as a small organic molecule, a polypeptide, an aptamer, an antibody or an intra-antibody,
- measuring the IL-10, TGFp 1 and/or FOXP3 activity involves determining a Ki on the IL-10, TGFp 1 and/or FOXP3 cloned and transfected in a stable manner into a CHO cell line, measuring cancer cells viability/survival, measuring cancer cell migration and invasion abilities, measuring cancer cell growth, measuring cancer cell proliferation, measuring cancer cell secretion in the presence or absence of the candidate compound.
- Tests and assays for screening and determining whether a candidate compound is a IL- 10, TGFp i and/or FOXP3 inhibitor, and/or FOXP3 modulator are well known in the art (Llorente et al, 2000; Clark et al, 2013; WO 2011/143280; US2013/0109619;
- WO2016196178 may be used to assess the potency and selectivity of the candidate compounds to inhibit IL-10, TGFp 1 and/or FOXP3 activity, or to modulate FOXP3 activity.
- Activities of the candidate compounds, their ability to bind IL-10, TGFp 1 and/or FOXP3 and their ability to inhibit IL-10, TGFp 1 and/or FOXP3 activity, modulate FOXP3 activity may be tested using isolated cancer cell, cancer cell lines or CHO cell line cloned and transfected in a stable manner by the human IL-10, TGFp i and/or FOXP3.
- FIGURES are a diagrammatic representation of FIGURES.
- FIG. 1 CLL-B cells express immune regulatory cytokines
- a and C Representative dot plots with the percentage of positive populations.
- C Representative co expression of IL10 and TGF 1 .
- B Representative co-expression of IL10 and FOXP3. The dotted line indicates that both sides are obtained from the same western blot. Wilcoxon signed-rank test, ns, not significant P >0.05.
- Figure 3 Phenotypic Characterization of CLL-B cells.
- Figure 4 Differential expression of regulatory factors between CD5 h,gh CD19 h,gh and CD5 d,m CD19 d,m subpopulations
- Figure 6 A polyfunctional score is correlated with factors at risk of progression
- IGHV Meat, M; Unmutated, UM
- ZAP70 and CD38 expressions -, Negative and +, positive
- Binet stage A and B or C
- %MTS Non responder, NR; Responder, R
- Isolated B and T CD4 + lymphocytes were isolated from total blood using Rosette B and CD4 + T isolation kits (STEMCELL). Isolated B and T cells subsets purity was assessed by flow cytometry analysis and was typically > 95%. Isolated B and T cells were cultured in RPMI 1640 containing L-glutamine and supplemented with 100 U/mg/ml penicillin/streptomycin (Life Technologies, USA), and 10% ECS (Biosera) at 37°C in humidified incubator containing 5% C02 at the concentration of 2xl0 6 /ml for 72 hours.
- B cells were stimulated or not with a combination of soluble CD40L (1 gg/ml; Miltenyi Biotec) and coated anti-IgM (20 gg/ml; Jackson ImmunoResearch) or a combination of soluble CD40L and IL21 (50ng/ml, Gibco by Life Technologies).
- CD4 + T cells were stimulated or not by coated anti-CD3 mAb (Hit-3a, 10 gg/ml; Ebioscience) and anti-CD28 mAb (CD28-2, 1 gg/ml; Ebio sciences).
- Lor cytokine detection by flow cytometry Brefeldin A (BE A) (10 gg/ml; Sigma Aldrich), PMA (500 ng/ml; Sigma Aldrich) and Ionomycin (1 gg/ml; Sigma Aldrich) were added for the last 4 hours of culture.
- BE A Brefeldin A
- PMA 500 ng/ml; Sigma Aldrich
- Ionomycin (1 gg/ml; Sigma Aldrich
- Sorted cells were lysed in NP40 lysis buffer (150 mM NaCl, 50 mM Tris-HCl, pH 7.4, 1 mM EDTA, 1% NP-40, 10% Glycerol with protease inhibitors). Proteins (2 to 20 pg) were separated on a 10% SDS-PAGE, transferred on a nitrocellulose membrane and incubated with rabbit monoclonal anti-LOXP3 antibody (D608R, Cell Signaling Technologies) and with anti- b actin mAb (AC-74, Sigma Aldrich), followed by appropriate secondary horseradish peroxidase-conjugated antibody. Detection was performed using ECL kit (Bio-Rad) and images acquired with a Chemidoc MP (BioRad).
- IL10, T ⁇ RbI and LOXP3 CLL expressing cells were first labeled for extracellular staining with anti-CD 19/V500 (HIB19), anti-CD5/V450 (UCHT2), anti-CD24/FITC (ML5), anti-CD25/APC-Cy7 (M-A251), anti-CD27/PerCP5.5 (M-T271) and anti-CD38/PE-Cy7 (HIT2) mAbs (BD Biosciences) for 20 min.
- cells were fixed (2% PFA in PBS IX), permeabilized (0.5 % saponin, 1% BSA in PBS IX) and further stained with anti- IL10/APC (JES3-19F1), anti-FOXP3/PE (259D/C7) or anti-TGFp i/PE (TW4-9E7) mAbs or with their respective isotypes (BD Biosciences) overnight at 4°C.
- anti-CD 19 A/500 HAB19
- CD5/FITC FITC
- CD25/APC-Cy7 M-A251
- CDl27/PE-Cy7 SB/199
- mAbs BD Biosciences.
- cells were fixed, permeabilized and stained with anti-IE 10/ APC (JES3-19F1), TGFp i/PE (TW4-9E7) or FOXP3/PE (259D/C7) (BD Biosciences).
- CD4 + T cells were labeled with CellTrace Violet (Invitrogen) immediately after isolation according to the manufacter’s protocol. After co-culture for 72 hours, cells were labeled with anti-CDl9/PEVk>770 (LT19), CD3/PerCPVk>700 (REA613) and CD5/APCVio770 (UCHT2), mAbs (Miltenyi biotec) for 20 min.
- cytofix/cytoperm buffer (BD biosciences) following the manufacturer’s protocol and then stained with anti-IFNy/PE (B27) (BD bioscience) and anti-TNFa/FITC (cA2) (Miltenyi biotech) or relevant isotype mAbs for 1 hour.
- Flow cytometry analysis was performed using a FACS Canto II driven by DIVA software (BD biosciences) and analyzed with the FlowJo software (Miltenyi Biotec).
- CLL-B cells express heterogeneous immunoregulatory cytokines profile.
- Capacity to secrete various immune modulatory cytokines was also quantified.
- TGFp i was secreted at low or high levels allowing the segregation of two groups.
- the amount of secreted TGFp i in the high secreting group was correlated to the fraction of expressing cells with levels similar to healthy controls ( Figure 1D and Data not shown). All the other cytokines tested were produced at low levels as compared to healthy controls.
- CLL-B cells constitutively express FOXP3.
- CLL-B cells could express, like Tregs, the major transcriptional regulator LOXP3.
- Llow cytometry analysis of peripheral viable CD5 + CDl9 + CLL-B cells revealed a LOXP3 + subpopulation present at highly variable extents among patient samples in culture for 3 days (ranging from 0.04 to 82%; mean 20.76 ⁇ 3.79%) (Ligure 2A and Data not shown).
- LOXP3 + cells were also observed in small needle aspirate of lymph node and at collection of peripheral blood samples (mean 5.68 ⁇ l . l9%).
- the memory CD5 + CDl9 + CD27 + CLL-B cells are the IL10 and/or TGFp 1 producing subsets.
- FOXP3 + cells express to a certain extent higher levels of CD5, CD19 and CD25 compared to FOXP3 cells.
- Analysis upon CD40/BCR triggering confirmed the results obtained with unstimulated cells in spite of a weaker CD5 detection ( Figure 3B).
- several other phenotypic markers usually associated to IL10 producing cells were not differentially expressed between IL-l0 +/ , TGFp i or FOXP3 +/ CLL-B cells and were not influenced by in vitro stimulation (Data not shown).
- CD5/CD19 expression levels discriminate intra-individual heterogeneity for regulatory-markers.
- CLL-B cells undergo a regulatory crosstalk with their T cell counterparts.
- CD5 hl CDl9 hl CLL-B cells also produce at variable extent the transcription factor FOXP3, a hallmark of Tregs.
- Co-cultures with autologous T cells have proved the functional regulatory implication of CLL-B cells in the induction of Tregs, the negative regulation of CD4 + T cell proliferation, altogether with a modulation of their pattern of secretion toward deficiency of tumor clearance.
- computation of the expression of the three regulatory factors in a polyfunctional score argues for its validation as a functional predictive marker of disease progression.
- CD5 + CD 19 + CD24 hi CD27 + and CD5 + CD 19 + CD24 hi CD38 hi subsets were able to produce IL-10 and TGFp i cytokines (Data not shown).
- Several other subtypes such as immature CD 19 + CD24 hi CD38 hi , CD 1 d + CD 19 + CD38 + IgM + CD 147 + GrB + or CDl lb + CDl9 + cells have also been described as IL10 competent cells (18, 19, 27, 32, 34).
- ILl0 + and TGFp i producing cells are memory CD5 + CDl9 + CD27 + that differ from the other described regulatory subtypes with markers such as CD25, CD24 and CD38.
- CLL-B cells also showed differential propensity to induce expression of TGFp i , an immunoregulatory factor for Tregs induction and CD8 + T cell anergy. These two immunomodulation have also been documented in CLL (11, 35, 36).
- TGF 1 expression when not already at the highest levels, was induced in a BCR/CD40 dependent manner, mimicking T/B cells interactions. We have previously shown that establishment of a threshold response to BCR triggering is mandatory for CLL-B cell heterogeneous survival (5).
- CLL-B cells and mainly CD5 hlgh CDl9 hlgh cells expressed also the transcription factor FOXP3 regardless of stimulation.
- FOXP3 is a novel indicator in CLL-B cells.
- a previous report described FOXP3 as present only in pre- apoptotic B cells in healthy controls (17). Therefore, we comforted our finding using different approaches including viability marker in flow cytometry and cell sorting to exclude any apoptotic cells or Tregs contamination that could account for FOXP3 expression.
- CD5 + CDl9 + samples we observed a strong match between the productions of the two immunoregulators FOXP3 and TGFp i .
- FOXP3 + B cells also expressed CD25, another key marker of Tregs, which has been observed in ILlO + Brl cells with antigen specific suppressive functions (37).
- a relationship between expression of CD5 and IL10 was already documented in CLL-B cells involving STAT3 and NFAT2 activation (38).
- This orientation might result from the secretion by CLL-B cells of the two main suppressive cytokines IL10 and TGFp i (26).
- This expansion was accompanied by a reduction of two major CD4 + T cell populations, e.g. expressing TNF-a and IFNy targets of regulatory B cells, tumor clearance and cytotoxic response (18, 19, 40).
- This suppressive mechanism was dependent on the ratio in the co-culture between T and B cells and reflecting the cellular distortion observed in CLL patients.
- CLL-B cells hindered or suppress, in a T/B ratio dependent manner again, CD4 + T cells division, a major hallmark of Bregs.
- CD5 hlgh CDl9 hlgh subpopulation as the functional subset expressing the highest levels of IL10, TGF 1 and FOXP3.
- This CD5 hlgh CDl9 hlgh subset allowed distinction of two groups of cases with differential evolution during patients follow up. A reasonable interpretation of this finding is that high levels of regulatory factors provoke exhaustion of tumor survey mechanisms and tumor escape. At the opposite, the lower levels seen in CD5 dim CDl9 dim do not generate warning signals toward tumor survey.
- TGF 1 only was statistically linked to ZAP70 expression or to BCR/CD40 triggering in IGHV unmutated patient samples arguing for the involvement of BCR signaling in the induction of TGFpU cells.
- induction of TGF 1 Bregs was reported upon stimulation with CTLA-4 by T cells (30).
- IL10 expressing cells were increased in IGHV mutated cases (21).
- our results indicate the presence of a constitutive pool of ILl0 + cells that was not induced upon B/T triggering.
- the polyfunctional score of the CD5 hlgh CDl9 hlgh subset correlated with two major indicators of B cell activation and antigenic recognition, e.g. IGHV mutational status and ZAP70 expression.
- higher polyfunctional score was associated with higher number of pejorative factors and patient disease progression necessitating treatment. With a measure of three different regulatory factors, the additional information given by the polyfunctional score might therefore become a better orientation indicator of disease progression and lack of tumor clearance.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Cell Biology (AREA)
- Chemical & Material Sciences (AREA)
- Urology & Nephrology (AREA)
- Biomedical Technology (AREA)
- Molecular Biology (AREA)
- Hematology (AREA)
- Microbiology (AREA)
- Biotechnology (AREA)
- Oncology (AREA)
- Hospice & Palliative Care (AREA)
- Food Science & Technology (AREA)
- Medicinal Chemistry (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- General Health & Medical Sciences (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
The present invention relates to methods for stratification and treatment of progressive chronic lymphocytic in a patient in need thereof Depending on the cytokines produced, several regulatory B cell (Breg) subsets contribute to immune resolution and their enrichment leads to disease-associated immunosuppression. Chronic Lymphocytic Leukemia (CLL) is a heterogeneous clonal B cell neoplasm ranging from indolent to rapidly progressive clinical course that eludes tumor clearance. This study presents a comprehensive phenotypic and functional analysis of the regulatory subpopulations in CLL patients at various stages of the disease. Cytokines profiling of CLL-B cells evidences the production at various extents of IL-10 and TGFβ1. Remarkably, CLL cells express also the FOXP3 transcription factor, an original marker of regulatory T cells. The three proteins are produced by subpopulations with markers of activated and memory Β cells defining a specific signature. Based on CD5 and CD19 expression, intraclonal heterogeneity showed differential regulatory factors production relevant to disease evolution. Functional studies proved their regulatory capacities targeting T cell differentiation, proliferation and secretion. IL-10, TGFβ1 and FOXP3 expressions combined in a polyfunctional score strongly correlated with high-risk factors of progression. This profiling helps to predict progression and pinpoints immune dysfunction in CLL. Thus, the present invention relates to methods for diagnosing progressive chronic lymphocytic in a patient comprising a step of determining the level of IL-10, TGFβ1 and FOXP3 and methods for treating said patient.
Description
METHODS FOR STRATIFICATION AND TREATMENT OF A PATIENT SUFFERING FROM CHRONIC FYMPHOCYTIC FEUKEMIA
FIEFD OF THE INVENTION:
The present invention relates to methods for stratification and treatment of progressive chronic lymphocytic in a patient in need thereof
BACKGROUND OF THE INVENTION:
Chronic lymphocytic leukemia (CLL) is a lymphoid malignancy prevalent in the elderly presenting with a heterogeneous clinical course. Some patients experience a progressive disease with rapid poor outcome while others exhibit an indolent leukemia that does not impact life expectancy (1). The disease results in the clonal expansion of small, mature B lymphocytes, which accumulate in the bone marrow, blood and secondary lymphoid organs. In progressive patients chemo- and immunotherapies are not curative and residual clonal cancer cells re- populate both lymphoid organs and peripheral blood; the antitumor immune surveillance being ineffective (1, 2). The heterogeneity of CLL has been investigated and related to immunophenotypic markers including CD5, CD 19 or CD38 expressions. Moreover, genetic studies have pointed out the importance of stereotyped CDR3 sequences of immunoglobulin heavy chain variable region (IGHV) genes and of antigen receptor (BCR)-triggered pathways leading to apoptotic defect (3, 4). These membrane and signaling features converge to an active antigen-driven selection in the clonal expansion of the malignant cells. Indeed, functional studies have confirmed the relevance of signals propagated by BCR triggering in the heterogeneity of leukemic cell survival in vitro. BCR effectors such as Spleen tyrosine kinase, Phospholipase C g2 or NFAT transcription factors are often overexpressed and/or constitutively activated in B-CLL cells and allow the expression of target genes important for cell survival (5, 6). Expression of unconventional activators, such as the zeta chain associated protein (ZAP-70) commonly expressed in T and NK cells, is also associated with increased cell survival. In addition, the capacity to generate and propagate such activating signals at the cellular level correlates with disease progression. By contrast, defective signal transduction is prevalent in indolent disease and related to an anergic phenotype of the leukemic cells (7, 8). The important driving force of BCR initiated signals is especially encountered in lymph nodes, which are important sites for antigen recognition and account for the accumulation of malignant cells in synergy with microenvironmental factors (9, 10).
Besides heterogeneous signaling capacity, the malignancy is also characterized by an imbalance of the immune subpopulations present in peripheral blood and lymphoid organs of CLL patients. T cell subsets that account for immune surveillance in tumor development encompassing CD4+ helpers (Th) and regulatory T cells (Tregs) have an altered ratio in these hematological malignancies (1 1 , 12). Thus, Tregs are increased in CLL and correlate with several clinical/biological features of progressive disease, whereas CD8+ T cells from CLL patients show functional defect in proliferation and cytotoxicity, but preserved cytokine production reflecting T-cell exhaustion (13). Such impairment of immunological homeostasis is often observed when ineffective antitumor immunity takes place during neoplasm progression and has been attributed to the production of several regulatory molecules and cytokines by specific B cell subsets ascribed to regulatory B cells (14-17). Recent data have shown that these B cell subsets play an important role in the direct or indirect suppression of inflammatory response and in the maintenance of tolerance through the production of IL10 (18- 21). Various teams have undertaken the phenotypic characterization of these subsets both in murine models and in human pathologies. However, this strategy did not identify a unique consensus but several phenotypes of progenitor populations with suppressive activity (16). Among others, murine CD5+ Bla and CDldhlghCD5+CDl9hlgh B10 cells are IL-10 producing cells (22, 23). Studies unraveling the functional properties of the CD5+ Bla lineage have uncovered regulatory properties leading to bias of the immune cells repertoire amongst which expansion of the Treg population and suppression of Thl and Thl7 differentiation (24). Human Breg subsets, identified by their capacity to suppress Thl differentiation and convert CD4+ T cells into Tregs via IL10 production, have also been described (25, 26). In peripheral blood from both healthy individuals and patients with autoimmune diseases or neoplasms, various IL10 producing subtypes have been reported (26). The latters include CDl9+CD24hlghCD38hlgh immature B cells, CD 19+CD24highCD27+ B10 cells,
CD 19+CD38+CD 1 d+IgM+CD 147+Granzyme+ B cells and CD27intermediateCD38high plasmablasts (19, 27, 28). Induction of IL10 in the various subsets seems to require signals from activated CD4+T cells with CD40L playing a major stimulatory role, while IL21 -dependant signals rather induce Granzyme (GrB) producing regulatory B cells preferentially (29). Additional induced Bregs can also exert a suppressive mechanism via the production of TGF 1 and indoleamine- 2,3 dioxygenase (IDO) (30).
Remarkably, the majority of these phenotypic features are commonly observed in B- CLL populations irrespective of their IGHV mutational status. Due to the expression of CD5, CLL-B cells have been hypothesized for long as being derived from a human Bl lineage
recognizing natural antibodies (31, 32). At present, CLL-B cells are considered as antigen- experienced B cells with an IGHV mutational status reflecting a T-dependent (mutated IGHV, M-IGHV) or T-independent (unmutated IGHV, UM-IGHV) memory phenotype with expression of CD27 in both cases (1). CLL malignant B cells have a clear survival advantage over the other normal B cells (33). Furthermore, diverse triggering events have been shown to induce IL10 in these cells, which share immunosuppressive capacities with B10 cells (21).
In the present invention, the inventors systematically analyzed the capacity for CLL cells from donors with indolent and progressive disease to produce in response to BCR/CD40 triggering key cytokines involved in immune modulation and in tumor surveillance. We found that a subset of leukemic cells of variable frequency produced constitutive IL10 and induced TGF 1 . Those cells also expressed the transcription factor FOXP3 that is a hallmark of Tregs. Cells that produce these three regulatory proteins display a specific signature somehow different from the already described B10 cells. Evidently, these cells share the regulatory properties attributed to Bregs toward CD4+ T cells. Finally, we give evidences that the combination of the three factors in a polyfunctional score is indicative of progression and might be used for the stratification of the disease. These novel findings provide also important insights on how CLL cells modify their immunological environment to their advantage.
SUMMARY OF THE INVENTION:
The present invention relates to methods for stratification and treatment of progressive chronic lymphocytic in a patient in need thereof.
In particular, the present invention is defined by the claims.
DETAILED DESCRIPTION OF THE INVENTION:
Depending on the cytokines produced, several regulatory B cell (Breg) subsets contribute to immune resolution and their enrichment leads to disease-associated immunosuppression. Chronic Lymphocytic Leukemia (CLL) is a heterogeneous clonal B cell neoplasm ranging from indolent to rapidly progressive clinical course that eludes tumor clearance. This study presents a comprehensive phenotypic and functional analysis of the regulatory subpopulations in CLL patients at various stages of the disease. Cytokines profiling of CLL-B cells evidences the production at various extents of IL-10 and TGFp 1 . Remarkably, CLL cells express also the FOXP3 transcription factor, an original marker of regulatory T cells. The three proteins are produced by subpopulations with markers of activated and memory B cells defining a specific signature. Based on CD5 and CD 19 expression, intraclonal heterogeneity showed differential regulatory factors production relevant to disease evolution. Functional studies proved their regulatory capacities targeting T cell differentiation,
proliferation and secretion. IL-10, TGFp i and FOXP3 expressions combined in a polyfunctional score strongly correlated with high-risk factors of progression. This profiling helps to predict progression and pinpoints immune dysfunction in CLL.
Accordingly, the invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment comprising a step of determining the level of IL-10, TGFp i and FOXP3 in a biological sample obtained from the patient.
The method of the invention may further comprises a step consisting of comparing the level of IL-10, TGFp i and FOXP3 in the biological sample with a reference value, wherein detecting differential in the level of IL-10, TGFp i and FOXP3 between the biological sample and the reference value is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
In some embodiments, the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of: i) determining the level of IL-10, TGFp i and FOXP3 in a biological sample obtained from the patient, ii) comparing the level determined at step i) with a reference value, wherein detecting differential in the level of IL-10, TGFp i and FOXP3 between the biological sample and the reference value is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
In some embodiments, the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
i) determining the level of IL-10, TGFp i and FOXP3 in a biological sample obtained from the patient at time tl,
ii) determining the level of IL-10, TGFp i and FOXP3 in a biological sample obtained from the patient at time t2, and
iii) an increase of the levels determined at step 1) and ii) between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
In some embodiments, the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
i) determining the level of IL-10, TGFp 1 and FOXP3 in a biological sample obtained from the patient at time tl,
ii) determining the level of IL-10, TGFp i and FOXP3 in a biological sample obtained from the patient at time t2,
iii) building a polyfunctional score combining the levels determined at step i) and a polyfunctional score combining the levels determined at step ii), and
iv) an increase of the polyfunctional score between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
In further aspect, the method of the invention comprises a step consisting of determining the level of IL-10, TGFp i and FOXP3 in the biological sample, followed by an unsupervised hierarchical cluster analysis and an index called polyfunctional score that was built by combining IL10, TGFp i and FOXP3 expressions. An increase of the polyfunctional score between two points of the clinical follow up is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
In some embodiments, the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment comprising the steps of: i) determining the expression level of IL-10, TGFp 1 and FOXP3 in a biological sample obtained from the patient, ii) applying an unsupervised hierarchical cluster analysis iii) building a polyfunctional score combining IL-10, TGFp i and FOXP3 expression levels determined at step i), an increase of the polyfunctional score value between two points of the clinical follow up is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
As used herein, the term“patient” denotes a mammal. Typically, a patient according to the invention refers to any patient (preferably human) afflicted or at risk to be afflicted with chronic lymphocytic leukemia. The term“patient” also refers to any patient (preferably human) afflicted or at risk to be afflicted with progressive chronic lymphocytic leukemia
As used herein, the term "chronic lymphocytic leukemia” or“CLL” has its general meaning in the art and refers to Acute myeloid leukemia such as revised in the World Health Organisation Classification C91.1. The term“Chronic lymphocytic leukemia” also refers to a heterogeneous clonal B cell neoplasm ranging from indolent to rapidly progressive clinical course that eludes tumor clearance. The term“Chronic lymphocytic leukemia” also refers to a lymphoid malignancy prevalent in the elderly presenting with a heterogeneous clinical course. Some patients experience a progressive disease with rapid poor outcome while others exhibit an indolent leukemia that does not impact life expectancy (1). The disease results in the clonal
expansion of small, mature B lymphocytes, which accumulate in the bone marrow, blood and secondary lymphoid organs.
As used herein the term“IL-10” has its general meaning in the art and refers to Interleukin 10.
As used herein the term“TGFp 1” has its general meaning in the art and refers to Transforming Growth Factor Beta 1.
As used herein the term“FOXP3” has its general meaning in the art and refers to Forkhead Box P3.
As used herein, the term“biological sample” refers to B cells, chronic lymphocytic leukemia B cells, bone marrow sample, lymphoid organ sample, blood sample, serum sample and plasma sample.
In a further aspect, the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of determining the level of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in a biological sample obtained from the patient.
The method of the invention may further comprises a step consisting of comparing the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 in the biological sample with a reference value, wherein detecting differential in the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 between the biological sample and the reference value is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
In some embodiment, the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
i) determining the level of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in a biological sample obtained from the patient,
ii) comparing the level determined at step i) with their corresponding predetermined reference value, and
iii) detecting differential between the level determined at step i) with the predetermined reference value is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
In some embodiments, the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
i) determining the level of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in a biological sample obtained from the patient at time tl,
ii) determining the level of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in a biological sample obtained from the patient at time t2, and
iii) an increase of the levels determined at step 1) and ii) between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
In some embodiments, the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
i) determining the level of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in a biological sample obtained from the patient at time tl,
ii) determining the level of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in a biological sample obtained from the patient at time t2,
iii) building a polyfunctional score combining the levels determined at step i) and a polyfunctional score combining the levels determined at step ii), and
iv) an increase of the polyfunctional score between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
In a further aspect, the method of the invention may further comprises a step consisting of combining the proportion of cells expressing IL-10, cells expressing TGFp i , and cells expressing FOXP3 in the biological sample to build a polyfunctional score. An increase of the polyfunctional score value between two points of the clinical follow up is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
In some embodiments, the present invention relates to a method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
i) determining the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 in a biological sample obtained from the patient,
ii) applying an unsupervised hierarchical cluster analysis,
iii) building a polyfunctional score combining the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 determined at step i). An increase of the polyfunctional score between two points of the clinical follow up is indicative of patient having
or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
In some embodiments, the cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 relates to cells expressing high level of IL-10, cells expressing high level of TGFp 1 , and cells expressing high level of FOXP3.
As used herein, the term“level of cells expressing a biomarker such as IL-10, TGFp 1 and FOXP3” has its general meaning in the art and refers to the quantity of cells expressing the biomarker or the number of cells expressing the biomarker. The term“level of cells expressing a biomarker” also refers to the density of cells expressing a biomarker. The term“level of cells expressing a biomarker” also refers to the percentage of cells expressing a biomarker. The term “level of cells expressing a biomarker” also refers to the level or percentage of B cells expressing a biomarker. The term“level of cells expressing a biomarker” also refers to the level or percentage of CLL B cells expressing a biomarker.
In some embodiments, the term“level of B cells expressing a biomarker” refers to the level or percentage of B cells expressing a biomarker compared to total B cells.
In some embodiments, the term“level of CLL B cells expressing a biomarker” refers to the level or percentage of CLL B cells expressing a bio marker compared to total CLL B cells.
As used herein, the“polyfunctional score” refers to a combined value of expression and the term“reference value” refers to a threshold value or a cut-off value. The setting of a single “polyfunctional score” or“reference value” thus allows discrimination between a progressive chronic lymphocytic leukemia patient and indolent chronic lymphocytic leukemia patient; discrimination between a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatments and a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia or not resistant to treatments; a poor and a good prognosis with respect to the aggressiveness, invasiveness and/or recurrence of chronic lymphocytic leukemia, cancer relapse and/or overall survival (OS) for a patient. Typically, a "threshold value", "cut-off value" or“polyfunctional score” can be determined experimentally, empirically, or theoretically. A threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative). Typically, the optimal sensitivity and specificity (and so the threshold value) can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data. Particularly, the person
skilled in the art may compare the quantity (obtained according to the method of the invention) with a defined threshold value. In one embodiment of the present invention, the threshold value is derived from the quantity (or ratio, or score) determined in a biological sample derived from one or more patients having chronic lymphocytic leukemia. Furthermore, retrospective measurement of the quantity (or ratio, or scores) in properly banked historical patient samples may be used in establishing these threshold values.
Predetermined reference values used for comparison may comprise “cut-off’ or “threshold” values that may be determined as described herein. Each reference (“cut-off’) or polyfunctional score value may be predetermined by carrying out a method comprising the steps of
a) providing a collection of samples (such as B cells, blood) from patients suffering of chronic lymphocytic leukemia;
b) determining the level of the bio markers or the level of cells expressing the bio markers for each sample contained in the collection provided at step a);
c) ranking the tumour samples according to said level;
d) classifying said samples in pairs of subsets of increasing, respectively decreasing, number of members ranked according to their level determined,
e) providing, for each sample provided at step a), information relating to the actual clinical outcome for the corresponding chronic lymphocytic leukemia patient (i.e. indolent chronic lymphocytic leukemia or progressive chronic lymphocytic leukemia; the duration of the event-free survival (EFS), metastasis- free survival (MFS) or the overall survival (OS) or both);
f) for each pair of subsets of samples, obtaining a Kaplan Meier percentage of survival curve;
g) for each pair of subsets of samples calculating the statistical significance (p value) between both subsets;
h) selecting as reference value for the quantity of cells, the value of the level the biomarkers or the level of cells for which the p value is the smallest.
For example, the level of the biomarkers or the level of cells expressing the biomarkers has been assessed for 100 cancer samples of 100 patients. The 100 samples are ranked according to their level of the biomarkers or their level of cells expressing the biomarkers. Sample 1 has the best level of the biomarkers or level of cells expressing the biomarkers and sample 100 has the worst level of the biomarkers or level of cells expressing the biomarkers. A first grouping provides two subsets: on one side sample Nr 1 and on the other side the 99 other samples. The
next grouping provides on one side samples 1 and 2 and on the other side the 98 remaining samples etc., until the last grouping: on one side samples 1 to 99 and on the other side sample Nr 100. According to the information relating to the actual clinical outcome for the corresponding chronic lymphocytic leukemia patient, Kaplan Meier curves are prepared for each of the 99 groups of two subsets. Also for each of the 99 groups, the p value between both subsets was calculated.
The reference value or polyfunctional score is selected such as the discrimination based on the criterion of the minimum p value is the strongest. In other terms, the level of the biomarkers or level of cells expressing the biomarkers corresponding to the boundary between both subsets for which the p value is minimum is considered as the reference value. It should be noted that the reference value is not necessarily the median value of levels of the m biomarkers or levels of cells expressing the biomarkers.
In routine work, the reference value (cut-off value) or polyfunctional score may be used in the present method to discriminate progressive chronic lymphocytic leukemia samples and therefore the corresponding patients.
Kaplan-Meier curves of percentage of survival as a function of time are commonly to measure the fraction of patients living for a certain amount of time after treatment and are well known by the man skilled in the art.
The man skilled in the art also understands that the same technique of assessment of the level of the bio markers or level of cells expressing the bio markers should of course be used for obtaining the reference value or polyfunctional score and thereafter for assessment of the level of the biomarkers or level of cells expressing the biomarkers of a patient subjected to the method of the invention.
In another embodiment, a score which is a composite of the level of the biomarkers or level of cells expressing the biomarkers may also be determined and compared to a reference value wherein a difference between said score and said reference value is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
In a particular embodiment, the score may be generated by a computer program.
In one embodiment, the reference value or the polyfunctional score may correspond to the level of the biomarkers or level of cells expressing the biomarkers determined in a sample associated with a patient having indolent chronic lymphocytic leukemia or a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia. Accordingly, a higher level of the biomarkers or level of cells expressing the biomarkers than the reference value or a highest level of the bio markers or level of cells expressing the bio markers in the
unsupervised hierarchical cluster analysis is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia, or resisting to treatments and a lower or equal level of the biomarkers or level of cells expressing the biomarkers than the reference value is indicative of a patient having indolent chronic lymphocytic leukemia or a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia.
In another embodiment, the reference value or the polyfunctional score may correspond to the level of the bio markers or level of cells expressing the bio markers determined in a sample associated with a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment. Accordingly, a higher or equal level of the biomarkers or level of cells expressing the biomarkers than the reference value or a highest level of the bio markers or level of cells expressing the bio markers in the unsupervised hierarchical cluster analysis is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment, and a lower level of the bio markers or level of cells expressing the bio markers than the reference value is indicative of a patient having indolent chronic lymphocytic leukemia or a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia.
In a further aspect, the method of the invention is performed in at least two times tl and t2. In some embodiments, the method of the invention is performed at time tl and at time t2 different from time tl .
Accordingly, an increase of the level of the biomarkers, level of cells expressing the biomarkers or the polyfunctional score is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment, and a stable or a decrease of the level of the biomarkers, level of cells expressing the biomarkers or the polyfunctional score is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
In some embodiments, the method of the invention further comprises determining the expression level of at least one biomarker selected from the group consisting of pro- inflammatory cytokines and/or regulatory cytokines.
In some embodiments, the method of the invention further comprises determining the level of cells (B cells or CLL B cells) expressing at least one bio marker selected from the group consisting of pro -inflammatory cytokines and/or regulatory cytokines.
The term“pro -inflammatory cytokines” has its general meaning in the art and refers to cytokines or soluble factors that are excreted from immune cells, such as specific helper T cells,
and that promote inflammation through pro-inflammatory signals such as IL 1 b, IL2, IL4 or IFNy.
The term“regulatory cytokines” has its general meaning in the art and refers to cytokines or soluble factors that secreted by cells involved in the remodeling of the immune response in normal or pathological environments such as IL6, IL8 or TNFa.
Accordingly, the equilibrium between positive and negative regulatory cytokines or level of cells expressing regulatory cytokines balancing toward a lower level of pro- inflammatory cytokines or level of cells expressing pro -inflammatory cytokines in favour of the negative regulatory cytokines is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
In some embodiments, a higher level of regulatory cytokines or level of cells expressing regulatory cytokines than the reference value and/or a lower level of pro -inflammatory cytokines or level of cells expressing pro -inflammatory cytokines than the reference value is indicative of a patient having or at risk of having or developing progressive chronic lymphocytic leukemia; and a lower level of regulatory cytokines or level of cells expressing regulatory cytokines than the reference value and/or a higher level of pro -inflammatory cytokines or level of cells expressing pro -inflammatory cytokines than the reference value is indicative of a patient having indolent chronic lymphocytic leukemia or a patient not having or not at risk of having or developing progressive chronic lymphocytic leukemia.
The level of cells expressing the bio markers is determined by any well-known method in the art. In some embodiments, the level of cells expressing the biomarkers is determined such as described in the example. In some embodiments, the level of cells expressing the biomarkers is determined by flow cytometry. In some embodiments, the level of cells expressing the biomarkers is determined by IHC or immunofluorescence.
For example, the quantification of the cells expressing the biomarkers is performed by contacting the biological sample with a binding partner (e.g. an antibody) specific for a cell biomarkers of said cells. Typically, the quantification of the cells expressing the biomarkers is performed by contacting the tissue tumour tissue sample with a binding partner (e.g. an antibody) specific for IL-10, TGFp 1 or FOXP3.
Typically, the level of cells expressing the bio markers is expressed as the percentage of the specific cells per total cells (set at 100%). In some embodiments, the level of cells expressing the biomarkers may also consist of the number of these cells that are counted per one unit of surface area, e.g. as the number of cells that are counted per mm2 of surface area of tumour tissue sample.
In some embodiments, the quantification of the cells expressing the biomarkers is performed by flow cytometry or Fluorescence-activated cell sorting (FACS). In some embodiments, the quantification of the cells expressing the bio markers is performed by flow cytometry such as described in the example.
The level of the biomarkers may be assessed by any of a wide variety of well-known methods for detecting expression of a transcribed nucleic acid or translated protein.
In one embodiment, the biomarker expression level is assessed by analyzing the expression of the protein translated from said gene. Said analysis can be assessed using an antibody (e.g., a radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled antibody), an antibody derivative (e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein of a protein/ligand pair (e.g., biotin-streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically to the protein translated from the gene encoding for the biomarker.
Methods for measuring the expression level of a biomarker in a sample may be assessed by any of a wide variety of well-known methods from one of skill in the art for detecting expression of a protein including, but not limited to, direct methods like mass spectrometry- based quantification methods, protein microarray methods, enzyme immunoassay (EIA), radioimmunoassay (RIA), Immunohistochemistry (IHC), Western blot analysis, ELISA, Luminex, ELISPOT and enzyme linked immunoabsorbant assay and undirect methods based on detecting expression of corresponding messenger ribonucleic acids (mRNAs). The mRNA expression profile may be determined by any technology known by a man skilled in the art. In particular, each mRNA expression level may be measured using any technology known by a man skilled in the art, including nucleic microarrays, quantitative Polymerase Chain Reaction (qPCR), next generation sequencing and hybridization with a labelled probe.
Said direct analysis can be assessed by contacting the sample with a binding partner capable of selectively interacting with the biomarker present in the sample. The binding partner may be an antibody that may be polyclonal or monoclonal, preferably monoclonal (e.g., a isotope-label, element-label, radio-labeled, chromophore- labeled, fluorophore-labeled, or enzyme-labeled antibody), an antibody derivative (e.g., an antibody conjugate with a substrate or with the protein or ligand of a protein of a protein/ligand pair (e.g., biotin-streptavidin)), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain, etc.) which binds specifically to the protein translated from the gene encoding for the biomarker of the invention. In another embodiment, the binding partner may be an aptamer.
The binding partners of the invention such as antibodies or aptamers, may be labelled with a detectable molecule or substance, such as an isotope, an element, a fluorescent molecule, a radioactive molecule or any others labels known in the art. Labels are known in the art that generally provide (either directly or indirectly) a signal.
As used herein, the term "labelled", with regard to the antibody, is intended to encompass direct labelling of the antibody or aptamer by coupling (i.e., physically linking) a detectable substance, such as an isotope, an element, a radioactive agent or a fluorophore (e.g. fluorescein isothiocyanate (FITC) or phycoerythrin (PE) or Indocyanine (Cy5)) to the antibody or aptamer, as well as indirect labelling of the probe or antibody by reactivity with a detectable substance. An antibody or aptamer of the invention may be produced with a specific isotope or a radioactive molecule by any method known in the art. For example radioactive molecules include but are not limited to radioactive atom for scintigraphic studies such as 1123, 1124, Inl l l, Rel86, Rel88, specific isotopes include but are not limited to 13C, 15N, 1261, 79Br, 8lBr.
The afore mentioned assays generally involve the binding of the binding partner (ie. antibody or aptamer) to a solid support. Solid supports which can be used in the practice of the invention include substrates such as nitrocellulose (e. g., in membrane or microtiter well form); polyvinylchloride (e. g., sheets or microtiter wells); polystyrene latex (e.g., beads or microtiter plates); polyvinylidene fluoride; diazotized paper; nylon membranes; activated beads, magnetically responsive beads, silicon wafers.
In a particular embodiment, an ELISA method can be used, wherein the wells of a microtiter plate are coated with a set of antibodies which recognize said biomarker. A sample containing or suspected of containing said bio marker is then added to the coated wells. After a period of incubation sufficient to allow the formation of antibody-antigen complexes, the plate(s) can be washed to remove unbound moieties and a detectably labelled secondary binding molecule added. The secondary binding molecule is allowed to react with any captured sample marker protein, the plate washed and the presence of the secondary binding molecule detected using methods well known in the art such as Singulex, Quanterix, MSD, Bioscale, Cytof.
In one embodiment, an Enzyme-linked immunospot (ELISpot) method may be used. Typically, the sample is transferred to a plate which has been coated with the desired anti biomarker capture antibodies. Revelation is carried out with biotinylated secondary Abs and standard colorimetric or fluorimetric detection methods such as streptavidin-alkaline phosphatase and NBT-BCIP and the spots counted.
In one embodiment, when multi-biomarker expression measurement is required, use of beads bearing binding partners of interest may be preferred. In a particular embodiment, the bead may be a cytometric bead for use in flow cytometry. Such beads may for example correspond to BD™ Cytometric Beads commercialized by BD Biosciences (San Jose, California). Typically, cytometric beads may be suitable for preparing a multiplexed bead assay. A multiplexed bead assay, such as, for example, the BD(TM) Cytometric Bead Array, is a series of spectrally discrete beads that can be used to capture and quantify soluble antigens. Typically, beads are labelled with one or more spectrally distinct fluorescent dyes, and detection is carried out using a multiplicity of photodetectors, one for each distinct dye to be detected. A number of methods of making and using sets of distinguishable beads have been described in the literature. These include beads distinguishable by size, wherein each size bead is coated with a different target-specific antibody (see e.g. Fulwyler and McHugh, 1990, Methods in Cell Biology 33:613-629), beads with two or more fluorescent dyes at varying concentrations, wherein the beads are identified by the levels of fluorescence dyes (see e.g. European Patent No. 0 126,450), and beads distinguishably labelled with two different dyes, wherein the beads are identified by separately measuring the fluorescence intensity of each of the dyes (see e.g. U.S. patent Nos. 4,499,052 and 4,717,655). Both one-dimensional and two-dimensional arrays for the simultaneous analysis of multiple antigens by flow cytometry are available commercially. Examples of one-dimensional arrays of singly dyed beads distinguishable by the level of fluorescence intensity include the BD(TM) Cytometric Bead Array (CBA) (BD Biosciences, San Jose, Calif.) and Cyto-Plex(TM) Flow Cytometry microspheres (Duke Scientific, Palo Alto, Calif.). An example of a two-dimensional array of beads distinguishable by a combination of fluorescence intensity (five levels) and size (two sizes) is the QuantumPlex(TM) microspheres (Bangs Laboratories, Fisher, Ind.). An example of a two- dimensional array of doubly-dyed beads distinguishable by the levels of fluorescence of each of the two dyes is described in Fulton et al. (1997, Clinical Chemistry 43(9): 1749-1756). The beads may be labelled with any fluorescent compound known in the art such as e.g. FITC (FL1), PE (FL2), fluorophores for use in the blue laser (e.g. PerCP, PE-Cy7, PE-Cy5, FL3 and APC or Cy5, FL4), fluorophores for use in the red, violet or UV laser (e.g. Pacific blue, pacific orange). In another particular embodiment, bead is a magnetic bead for use in magnetic separation. Magnetic beads are known to those of skill in the art. Typically, the magnetic bead is preferably made of a magnetic material selected from the group consisting of metals (e.g. ferrum, cobalt and nickel), an alloy thereof and an oxide thereof. In another particular embodiment, bead is bead that is dyed and magnetized.
In one embodiment, protein microarray methods may be used. Typically, at least one antibody or aptamer directed against the biomarker is immobilized or grafted to an array(s), a solid or semi- so lid surface(s). A sample containing or suspected of containing the biomarker is then labelled with at least one isotope or one element or one fluorophore or one colorimetric tag that are not naturally contained in the tested sample. After a period of incubation of said sample with the array sufficient to allow the formation of antibody-antigen complexes, the array is then washed and dried. After all, quantifying said biomarker may be achieved using any appropriate microarray scanner like fluorescence scanner, colorimetric scanner, SIMS (secondary ions mass spectrometry) scanner, maldi scanner, electromagnetic scanner or any technique allowing to quantify said labels.
In another embodiment, the antibody or aptamer grafted on the array is labelled.
In another embodiment, reverse phase arrays may be used. Typically, at least one sample is immobilized or grafted to an array(s), a solid or semi- so lid surface(s). An antibody or aptamer against the suspected biomarker is then labelled with at least one isotope or one element or one fluorophore or one colorimetric tag that are not naturally contained in the tested sample. After a period of incubation of said antibody or aptamer with the array sufficient to allow the formation of antibody-antigen complexes, the array is then washed and dried. After all, detecting quantifying and counting by D-SIMS said biomarker containing said isotope or group of isotopes, and a reference natural element, and then calculating the isotopic ratio between the biomarker and the reference natural element may be achieved using any appropriate microarray scanner like fluorescence scanner, colorimetric scanner, SIMS (secondary ions mass spectrometry) scanner, maldi scanner, electromagnetic scanner or any technique allowing to quantify said labels.
In one embodiment, said direct analysis can also be assessed by mass Spectrometry. Mass spectrometry-based quantification methods may be performed using either labelled or unlabelled approaches (DeSouza and Siu, 2012). Mass spectrometry-based quantification methods may be performed using chemical labeling, metabolic labelingor proteolytic labeling. Mass spectrometry-based quantification methods may be performed using mass spectrometry label free quantification, LTQ Orbitrap Velos, LTQ-MS/MS, a quantification based on extracted ion chromatogram EIC (progenesis LC-MS, Liquid chromatography-mass spectrometry) and then profile alignment to determine differential expression of the biomarker.
In another embodiment, the biomarker expression level is assessed by analyzing the expression of mRNA transcript or mRNA precursors, such as nascent RNA, of biomarker gene. Said analysis can be assessed by preparing mRNA/cDNA from cells in a sample from a subject,
and hybridizing the mRNA/cDNA with a reference polynucleotide. The prepared mRNA/cDNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, polymerase chain reaction analyses, such as quantitative PCR (TaqMan), and probes arrays such as GeneChip(TM) DNA Arrays ( AFF YMETRIX) .
Advantageously, the analysis of the expression level of mRNA transcribed from the gene encoding for biomarkers involves the process of nucleic acid amplification, e. g., by RT- PCR (the experimental embodiment set forth in U. S. Patent No. 4,683, 202), ligase chain reaction (Barany, 1991), self- sustained sequence replication (Guatelli et al., 1990), transcriptional amplification system (Kwoh et al., 1989), Q-Beta Replicase (Lizardi et al., 1988), rolling circle replication (U. S. Patent No. 5,854, 033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers. As used herein, amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5' or 3' regions of a gene (plus and minus strands, respectively, or vice-versa) and contain a short region in between. In general, amplification primers are from about 10 to 30 nucleotides in length and flank a region from about 50 to 200 nucleotides in length. Under appropriate conditions and with appropriate reagents, such primers permit the amplification of a nucleic acid molecule comprising the nucleotide sequence flanked by the primers.
In a further aspect, the method of the present invention is suitable for monitoring chronic lymphocytic leukemia progression in a patient in need thereof.
In a further aspect, the method of the present invention is suitable for determining whether a patient is eligible or not to an anti-cancer treatment or an anti-cancer therapy. For example, when it is determined that the patient is having or at risk of having or developing progressive chronic lymphocytic leukemia then the physician can take the choice to administer the patient with an anti-cancer treatment. Typically, the treatment includes chemotherapy, radiotherapy, radio immunotherapy, immunotherapy and drugs suitable for the treatment and prevention of progressive chronic lymphocytic leukemia.
The term“anti-cancer treatment” or“anti-cancer therapy” has its general meaning in the art and refers to anti-cancer compounds used in anti-cancer therapy such as tyrosine kinase inhibitors, tyrosine kinase receptor (TKR) inhibitors, EGFR tyrosine kinase inhibitors, anti- EGFR compounds, anti-HER2 compounds, Vascular Endothelial Growth Factor Receptors (VEGFRs) pathway inhibitors, interferon therapy, alkylating agents, anti-metabolites,
immunotherapeutic agents such as sipuleucel-T, Androgen deprivation therapy (ADT), Interferons (IFNs), Interleukins, radiotherapeutic agents (such as Ra223, Pb2l2) and chemotherapeutic agents such as described below.
The term“tyrosine kinase inhibitor” or“TKΊ” has its general meaning in the art and refers to any of a variety of therapeutic agents or drugs such as compounds inhibiting tyrosine kinase, tyrosine kinase receptor inhibitors (TKRI), EGFR tyrosine kinase inhibitors, EGFR antagonists. The term“tyrosine kinase inhibitor” or“TKI” has its general meaning in the art and refers to any of a variety of therapeutic agents or drugs that act as selective or non-selective inhibitors of receptor and/or non-receptor tyrosine kinases. Tyrosine kinase inhibitors and related compounds are well known in the art and described in U.S Patent Publication 2007/0254295, which is incorporated by reference herein in its entirety. It will be appreciated by one of skill in the art that a compound related to a tyrosine kinase inhibitor will recapitulate the effect of the tyrosine kinase inhibitor, e.g., the related compound will act on a different member of the tyrosine kinase signaling pathway to produce the same effect as would a tyrosine kinase inhibitor of that tyrosine kinase. Examples of tyrosine kinase inhibitors and related compounds suitable for use in methods of embodiments of the present invention include, but are not limited to Erlotinib, sunitinib (Sutent; SU11248), dasatinib (BMS-354825), PP2, BEZ235, saracatinib, gefitinib (Iressa), erlotinib (Tarceva; OSI-1774), lapatinib (GW572016; GW2016), canertinib (Cl 1033), semaxinib (SU5416), vatalanib (PTK787/ZK222584), sorafenib (BAY 43-9006), imatinib (Gleevec; STI571), leflunomide (SU101), vandetanib (Zactima; ZD6474), MK-2206 (8-[4-aminocyclobutyl)phenyl]-9-phenyl-l,2,4-triazolo[3,4- f][l,6]naphthyridin-3(2H)-one hydrochloride) derivatives thereof, analogs thereof, and combinations thereof. Additional tyrosine kinase inhibitors and related compounds suitable for use in the present invention are described in, for example, U.S Patent Publication 2007/0254295, U.S. Pat. Nos. 5,618,829, 5,639,757, 5,728,868, 5,804,396, 6,100,254, 6,127,374, 6,245,759, 6,306,874, 6,313,138, 6,316,444, 6,329,380, 6,344,459, 6,420,382, 6,479,512, 6,498,165, 6,544,988, 6,562,818, 6,586,423, 6,586,424, 6,740,665, 6,794,393,
6,875,767, 6,927,293, and 6,958,340, all of which are incorporated by reference herein in their entirety. In certain embodiments, the tyrosine kinase inhibitor is a small molecule kinase inhibitor that has been orally administered and that has been the subject of at least one Phase I clinical trial, more particularly at least one Phase II clinical, even more particularly at least one Phase III clinical trial, and most particularly approved by the FDA for at least one hematological or oncological indication. Examples of such inhibitors include, but are not limited to Erlotinib, Gefitinib, Lapatinib, Canertinib, BMS-599626 (AC-480), Neratinib, KRN-633, CEP-11981,
Imatinib, Nilotinib, Dasatinib, AZM-475271, CP-724714, TAK-165, Sunitinib, Vatalanib, CP- 547632, Vandetanib, Bosutinib, Lestaurtinib, Tandutinib, Midostaurin, Enzastaurin, AEE-788, Pazopanib, Axitinib, Motasenib, OSI-930, Cediranib, KRN-951, Dovitinib, Seliciclib, SNS- 032, PD-0332991, MKC-I (Ro-3 l7453; R-440), Sorafenib, ABT-869, Brivanib (BMS- 582664), SU-14813, Telatinib, SU-6668, (TSU-68), L-21649, MLN-8054, AEW-541, and PD- 0325901.
EGFR tyrosine kinase inhibitors as used herein include, but are not limited to compounds selected from the group consisting of but not limited to Erlotinib, lapatinib, Rociletinib (CO- 1686), gefitinib, Dacomitinib (PF-00299804), Afatanib, Brigatinib (AP26113), WJTOG3405, NEJ002, AZD9291, HM61713, EGF816, ASP 8273, AC 0010. Examples of antibody EGFR inhibitors include Cetuximab, panitumumab, matuzumab, zalutumumab, nimotuzumab, necitumumab, Imgatuzumab (GA201, RO5083945), and ABT- 806.
The term "chemotherapeutic agent" has its general meaning in the art and refers to chemical compounds that are effective in inhibiting tumor growth. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolo melamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CBI-TMI); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlomaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as the enediyne antibiotics (e.g. calicheamicin, especially calicheamicin (11 and calicheamicin 211, see, e.g., Agnew Chem Intl. Ed. Engl. 33: 183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including
morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idanrbicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptomgrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, 5-FU; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defo famine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK®; razoxane; rhizoxin; sizofiran; spirogennanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridinA and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobromtol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g. paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.].) and doxetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-l l ; topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included in this definition are antihormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
The term“anti-cancer treatment” or“anti-cancer therapy” also refers to targeted cancer therapy. Targeted cancer therapies are drugs or other substances that block the growth and
spread of cancer by interfering with specific molecules ("molecular targets") that are involved in the growth, progression, and spread of cancer. Targeted cancer therapies are sometimes called "molecularly targeted drugs", "molecularly targeted therapies", "precision medicines", or similar names. In some embodiments, the targeted therapy consists of administering the subject with a tyrosine kinase inhibitor as defined above.
The term “anti-cancer treatment” or “anti-cancer therapy” also refers to immunotherapeutic agent. The term "immunotherapeutic agent" as used herein, refers to a compound, composition or treatment that indirectly or directly enhances, stimulates or increases the body's immune response against cancer cells and/or that decreases the side effects of other anticancer therapies. Immunotherapy is thus a therapy that directly or indirectly stimulates or enhances the immune system's responses to cancer cells and/or lessens the side effects that may have been caused by other anti-cancer agents. Immunotherapy is also referred to in the art as immunologic therapy, biological therapy biological response modifier therapy and biotherapy. Examples of common immunotherapeutic agents known in the art include, but are not limited to, cytokines, cancer vaccines, monoclonal antibodies and non-cytokine adjuvants. Alternatively, the immunotherapeutic treatment may consist of administering the subject with an amount of immune cells (T cells, NK, cells, dendritic cells, B cells...). Immunotherapeutic agents can be non-specific, i.e. boost the immune system generally so that the human body becomes more effective in fighting the growth and/or spread of cancer cells, or they can be specific, i.e. targeted to the cancer cells themselves immunotherapy regimens may combine the use of non-specific and specific immunotherapeutic agents. Non-specific immunotherapeutic agents are substances that stimulate or indirectly improve the immune system. Non-specific immunotherapeutic agents have been used alone as a main therapy for the treatment of cancer, as well as in addition to a main therapy, in which case the non-specific immunotherapeutic agent functions as an adjuvant to enhance the effectiveness of other therapies (e.g. cancer vaccines). Non-specific immunotherapeutic agents can also function in this latter context to reduce the side effects of other therapies, for example, bone marrow suppression induced by certain chemotherapeutic agents. Non-specific immunotherapeutic agents can act on key immune system cells and cause secondary responses, such as increased production of cytokines and immunoglobulins. Alternatively, the agents can themselves comprise cytokines. Non specific immunotherapeutic agents are generally classified as cytokines or non-cytokine adjuvants. A number of cytokines have found application in the treatment of cancer either as general non-specific immunotherapies designed to boost the immune system, or as adjuvants provided with other therapies. Suitable cytokines include, but are not limited to, interferons,
interleukins and colony- stimulating factors. Interferons (IFNs) contemplated by the present invention include the common types of IFNs, IFN-alpha (IFN-a), and IFN-beta (IFN-b). IFNs can act directly on cancer cells, for example, by slowing their growth, promoting their development into cells with more normal behaviour and/or increasing their production of antigens thus making the cancer cells easier for the immune system to recognise and destroy. IFNs can also act indirectly on cancer cells, for example, by slowing down angiogenesis, boosting the immune system and/or stimulating natural killer (NK) cells, T cells and macrophages. Recombinant IFN-alpha is available commercially as Roferon (Roche Pharmaceuticals) and Intron A (Schering Corporation). Interleukins contemplated by the present invention include IL-2, IL-4, IL-l l and IL-12. Examples of commercially available recombinant interleukins include Proleukin® (IL-2; Chiron Corporation) and Neumega® (IL- 12; Wyeth Pharmaceuticals). Zymogenetics, Inc. (Seattle, Wash.) is currently testing a recombinant form of IL-21, which is also contemplated for use in the combinations of the present invention. Colony-stimulating factors (CSLs) contemplated by the present invention include sargramostim. Treatment with one or more growth factors can help to stimulate the generation of new blood cells in subjects undergoing traditional chemotherapy. Accordingly, treatment with CSLs can be helpful in decreasing the side effects associated with chemotherapy and can allow for higher doses of chemotherapeutic agents to be used. In addition to having specific or non-specific targets, immunotherapeutic agents can be active, i.e. stimulate the body's own immune response, or they can be passive, i.e. comprise immune system components that were generated external to the body. Passive specific immunotherapy typically involves the use of one or more monoclonal antibodies that are specific for a particular antigen found on the surface of a cancer cell or that are specific for a particular cell growth factor. Monoclonal antibodies may be used in the treatment of cancer in a number of ways, for example, to enhance a subject’s immune response to a specific type of cancer, to interfere with the growth of cancer cells by targeting specific cell growth factors, such as those involved in angiogenesis, or by enhancing the delivery of other anticancer agents to cancer cells when linked or conjugated to agents such as chemotherapeutic agents, radioactive particles or toxins. Monoclonal antibodies currently used as cancer immunotherapeutic agents that are suitable for inclusion in the combinations of the present invention include, but are not limited to, rituximab (Rituxan®), trastuzumab (Herceptin®), ibritumomab tiuxetan (Zevalin®), tositumomab (Bexxar®), cetuximab (C-225, Erbitux®), bevacizumab (Avastin®), gemtuzumab ozogamicin (Mylotarg®), alemtuzumab (Campath®), and BL22. Other examples include anti-CTLA4 antibodies (e.g. Ipilimumab), anti-PDl antibodies, anti-PDLl antibodies, anti-TIMP3
antibodies, anti-LAG3 antibodies, anti-B7H3 antibodies, anti-B7H4 antibodies or anti-B7H6 antibodies. In some embodiments, antibodies include B cell depleting antibodies. Typical B cell depleting antibodies include but are not limited to anti-CD20 monoclonal antibodies [e.g. Rituximab (Roche), Ibritumomab tiuxetan (Bayer Schering), Tositumomab (GlaxoSmithKline), AME-l33v (Applied Molecular Evolution), Ocrelizumab (Roche), Ofatumumab (HuMax-CD20, Gemnab), TRU-015 (Trubion) and IMMU-106 (Immuno medics)], an anti-CD22 antibody [e.g. Epratuzumab, Leonard et al, Clinical Cancer Research (Z004) 10: 53Z7-5334], anti-CD79a antibodies, anti-CD27 antibodies, or anti-CDl9 antibodies (e.g. U.S. Pat. No. 7,109,304), anti-BAFF-R antibodies (e.g. Belimumab, GlaxoSmithKline), anti- APRIL antibodies (e.g. anti-human APRIL antibody, ProSci inc.), and anti-IL-6 antibodies [e.g. previously described by De Benedetti et al, J Immunol (2001) 166: 4334-4340 and by Suzuki et al, Europ J of Immunol (1992) 22 (8) 1989-1993, fully incorporated herein by reference]. The immunotherapeutic treatment may consist of allografting, in particular, allograft with hematopoietic stem cell HSC. The immunotherapeutic treatment may also consist in an adoptive immunotherapy as described by Nicholas P. Restifo, Mark E. Dudley and Steven A. Rosenberg“Adoptive immunotherapy for cancer: harnessing the T cell response, Nature Reviews Immunology, Volume 12, April 2012). In adoptive immunotherapy, the subject’s circulating lymphocytes, NK cells, are isolated amplified in vitro and readministered to the subject. The activated lymphocytes or NK cells are most particularly be the subject’s own cells that were earlier isolated from a blood or tumor sample and activated (or“expanded”) in vitro.
The term“anti-cancer treatment” or“anti-cancer therapy” also refers to BRAF inhibitors such as vemurafenib, dacarbazine, dabrafenib, BMS-908662, LGX818, PLX3603, RAF265, R05185426, GSK2118436 and compounds described in Morris and Kopetz, 2013.
The term“anti-cancer treatment” or“anti-cancer therapy” also refers to radiotherapeutic agent. The term "radiotherapeutic agent" as used herein, is intended to refer to any radiotherapeutic agent known to one of skill in the art to be effective to treat or ameliorate cancer, without limitation. For instance, the radiotherapeutic agent can be an agent such as those administered in brachytherapy or radionuclide therapy such as Ra223 or Pb2l2. Such methods can optionally further comprise the administration of one or more additional cancer therapies, such as, but not limited to, chemotherapies, and/or another radiotherapy.
In one embodiment, said active compounds may be contained in the same composition or administrated separately.
In a further aspect, the present invention relates to a compound selected from the group consisting of IL-10 inhibitor, TGFp i inhibitor and/or FOXP3 modulator for use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof
In some embodiments, the present invention relates to a compound selected from the group consisting of IL-10 inhibitor, TGFp i inhibitor and/or FOXP3 modulator for use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof wherein the patient was being classified as having or at risk of having or developing progressive chronic lymphocytic leukemia by the method as above described.
In some embodiments, the invention relates to combined preparation of the compounds of the invention for simultaneous, separate or sequential use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof
Typically, 1, 2 or 3 compounds selected from the group consisting of IL-10 inhibitor, TGFp 1 inhibitor and FOXP3 modulator is used according to the invention.
In some embodiments, the present invention relates to an IL-10 inhibitor for use according to the invention.
In some embodiments, the present invention relates to a TGFp i inhibitor for use according to the invention.
In some embodiments, the present invention relates to a FOXP3 modulator for use according to the invention.
In some embodiments, the present invention relates to an IL-10 inhibitor and TGFp 1 inhibitor for use according to the invention.
In some embodiments, the present invention relates to an IL-10 inhibitor and FOXP3 modulator for use according to the invention.
In some embodiments, the present invention relates to a TGFp i inhibitor and FOXP3 modulator for use according to the invention.
In some embodiments, the present invention relates to an IL-10 inhibitor, TGFp 1 inhibitor and FOXP3 modulator for use according to the invention.
As used herein, the term "treatment" or "treat" refer to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of subjects at risk of contracting the disease or suspected to have contracted the disease as well as subjects who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse. The treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or
recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment. By "therapeutic regimen" is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy. A therapeutic regimen may include an induction regimen and a maintenance regimen. The phrase "induction regimen" or "induction period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease. The general goal of an induction regimen is to provide a high level of drug to a subject during the initial period of a treatment regimen. An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both. The phrase "maintenance regimen" or "maintenance period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a subject during treatment of an illness, e.g., to keep the subject in remission for long periods of time (months or years). A maintenance regimen may employ continuous therapy (e.g., administering a drug at regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
The term“modulator” has its general meaning in the art and refers to a target inhibitor or activator. The term“modulator” also refers to a compound that increase or decrease the expression of a specific gene.
The term“inhibitor” has its general meaning in the art and refers to a compound that selectively blocks or inactivates the target (IL-10, TGFp i and/or FOXP3). The term“inhibitor” also refers to a compound that selectively blocks the binding of the target to its substrate. The term“inhibitor” also refers to a compound able to prevent the action of the target for example by inhibiting the target controls of downstream effectors such as inhibiting the activation of the target pathway signalling. As used herein, the term“selectively blocks or inactivates” refers to a compound that preferentially binds to and blocks or inactivates the target with a greater affinity and potency, respectively, than its interaction with the other sub-types of the target family. Compounds that block or inactivate the target, but that may also block or inactivate other target sub-types, as partial or full inhibitors, are contemplated. The term“inhibitor” also refers to a compound that inhibits the target expression. Typically, an inhibitor is a small organic molecule, a polypeptide, an aptamer, an antibody, an oligonucleotide or a ribozyme.
Tests and assays for determining whether a compound is a modulator, an inhibitor or an activator are well known by the skilled person in the art such as described in (Llorente et al, 2000; Clark et al, 2013; WO 2011/143280; US2013/0109619; WO2016196178; WO 2015/124715; US 2009/0325868).
The term“activator” has its general meaning in the art and refers to any compound that can directly or indirectly stimulate the signal transduction cascade related to the target (FOXP3). The term“activator” also refers to a compound that selectively activates the target. Typically, a FOXP3 activator is a small organic molecule, a peptide, a modified FOXP3 or an activator of FOXP3 expression.
The term“expression” when used in the context of expression of a gene or nucleic acid refers to the conversion of the information, contained in a gene, into a gene product. A gene product can be the direct transcriptional product of a gene (e.g., mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA or any other type of RNA) or a protein produced by translation of a mRNA. Gene products also include messenger RNAs, which are modified, by processes such as capping, polyadenylation, methylation, and editing, and proteins modified by, for example, methylation, acetylation, phosphorylation, ubiquitination, SUMOylation, ADP- ribosylation, myristilation, and glycosylation.
An“inhibitor of expression” refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene.
An“activator of expression” refers to a natural or synthetic compound that has a biological effect to activate the expression of a gene.
The term“IL-10 inhibitor” has its general meaning in the art and refers to compounds such as B-N10; ABF 13; B-S10; anti-IL-lO monoclonal antibody; IL-10 receptor antagonists; IL-10 receptor expression inhibitors and compounds described in Llorente et al, 2000; Clark et al, 2013; WO2011/143280; US2013/0109619; WO2016196178.
The term“TGFP inhibitor” has its general meaning in the art and refers to compounds such as Galunisertib (LY2157299); TEW-7197; LY3022859; IMC-TRI; Fresolimumab (GC- 1008); PF-03446962; Trabedersen (AP- 12009); Belagenpumatucel-L; Pirfenidone; and compounds described in Herbertz et al, 2015; W02010/089443; US2011/0294734; US2012/0315256; US2007/0014767; US2013/0225655.
The term “FOXP3 modulator” has its general meaning in the art and refers to compounds such as histone/protein deacetylases (HDAC), histone/protein deacetylases inhibitors (HDACi) and polypeptides and compounds described in US2010/0034786; WO2013/050596; Casares et al., 2010; Lozano et al., 2017.
In another embodiment, the compound of the invention is an aptamer. Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition. Aptamers are oligonucleotide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity. Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., 1990. The random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence. Possible modifications, uses and advantages of this class of molecules have been reviewed in Jayasena S.D., 1999. Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al., 1996). Then after raising aptamers directed against the target of the invention as above described, the skilled man in the art can easily select those blocking or inactivating the target.
In another embodiment, the compound of the invention is an antibody (the term including“antibody portion”) directed against the target and which is an IL-10 inhibitor, TGFp 1 inhibitor or a FOXP3 modulator.
In one embodiment of the antibodies or portions thereof described herein, the antibody is a monoclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a polyclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a humanized antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a chimeric antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a light chain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a heavy chain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fab portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a F(ab')2 portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fc portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fv portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a variable domain of the antibody. In one embodiment of the
antibodies or portions thereof described herein, the portion of the antibody comprises one or more CDR domains of the antibody.
As used herein, "antibody" includes both naturally occurring and non-naturally occurring antibodies. Specifically, "antibody" includes polyclonal and monoclonal antibodies, and monovalent and divalent fragments thereof. Furthermore, "antibody" includes chimeric antibodies, wholly synthetic antibodies, single chain antibodies, and fragments thereof. The antibody may be a human or nonhuman antibody. A nonhuman antibody may be humanized by recombinant methods to reduce its immunogenicity in man.
Antibodies are prepared according to conventional methodology. Monoclonal antibodies may be generated using the method of Kohler and Milstein (Nature, 256:495, 1975). To prepare monoclonal antibodies useful in the invention, a mouse or other appropriate host animal is immunized at suitable intervals (e.g., twice-weekly, weekly, twice-monthly or monthly) with antigenic forms of the target. The animal may be administered a final "boost" of antigen within one week of sacrifice. It is often desirable to use an immunologic adjuvant during immunization. Suitable immunologic adjuvants include Freund's complete adjuvant, Freund's incomplete adjuvant, alum, Ribi adjuvant, Hunter's Titermax, saponin adjuvants such as QS21 or Quil A, or CpG-containing immunostimulatory oligonucleotides. Other suitable adjuvants are well-known in the field. The animals may be immunized by subcutaneous, intraperitoneal, intramuscular, intravenous, intranasal or other routes. A given animal may be immunized with multiple forms of the antigen by multiple routes.
Briefly, the antigen may be provided as synthetic peptides corresponding to antigenic regions of interest in the target. Following the immunization regimen, lymphocytes are isolated from the spleen, lymph node or other organ of the animal and fused with a suitable myeloma cell line using an agent such as polyethylene glycol to form a hydridoma. Following fusion, cells are placed in media permissive for growth of hybridomas but not the fusion partners using standard methods, as described (Coding, Monoclonal Antibodies: Principles and Practice: Production and Application of Monoclonal Antibodies in Cell Biology, Biochemistry and Immunology, 3rd edition, Academic Press, New York, 1996). Following culture of the hybridomas, cell supernatants are analyzed for the presence of antibodies of the desired specificity, i.e., that selectively bind the antigen. Suitable analytical techniques include ELISA, flow cytometry, immunoprecipitation, and western blotting. Other screening techniques are well-known in the field. Preferred techniques are those that confirm binding of antibodies to conformationally intact, natively folded antigen, such as non-denaturing ELISA, flow cytometry, and immunoprecipitation.
Significantly, as is well-known in the art, only a small portion of an antibody molecule, the paratope, is involved in the binding of the antibody to its epitope (see, in general, Clark, W. R. (1986) The Experimental Foundations of Modem Immunology Wiley & Sons, Inc., New York; Roitt, I. (1991) Essential Immunology, 7th Ed., Blackwell Scientific Publications, Oxford). The Fc' and Fc regions, for example, are effectors of the complement cascade but are not involved in antigen binding. An antibody from which the pFc' region has been enzymatically cleaved, or which has been produced without the pFc' region, designated an F(ab’)2 fragment, retains both of the antigen binding sites of an intact antibody. Similarly, an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region, designated a Fab fragment, retains one of the antigen binding sites of an intact antibody molecule. Proceeding further, Fab fragments consist of a covalently bound antibody light chain and a portion of the antibody heavy chain denoted Fd. The Fd fragments are the major determinant of antibody specificity (a single Fd fragment may be associated with up to ten different light chains without altering antibody specificity) and Fd fragments retain epitope-binding ability in isolation.
Within the antigen-binding portion of an antibody, as is well-known in the art, there are complementarity determining regions (CDRs), which directly interact with the epitope of the antigen, and framework regions (FRs), which maintain the tertiary structure of the paratope (see, in general, Clark, 1986; Roitt, 1991). In both the heavy chain Fd fragment and the light chain of IgG immunoglobulins, there are four framework regions (FR1 through FR4) separated respectively by three complementarity determining regions (CDR1 through CDRS). The CDRs, and in particular the CDRS regions, and more particularly the heavy chain CDRS, are largely responsible for antibody specificity.
It is now well-established in the art that the non-CDR regions of a mammalian antibody may be replaced with similar regions of conspecific or heterospecific antibodies while retaining the epitopic specificity of the original antibody. This is most clearly manifested in the development and use of "humanized" antibodies in which non-human CDRs are covalently joined to human FR and/or Fc/pFc’ regions to produce a functional antibody.
This invention provides in certain embodiments compositions and methods that include humanized forms of antibodies. As used herein, "humanized" describes antibodies wherein some, most or all of the amino acids outside the CDR regions are replaced with corresponding amino acids derived from human immunoglobulin molecules. Methods of humanization include, but are not limited to, those described in U.S. Pat. Nos. 4,816,567, 5,225,539, 5,585,089, 5,693,761, 5,693,762 and 5,859,205, which are hereby incorporated by reference.
The above U.S. Pat. Nos. 5,585,089 and 5,693,761, and WO 90/07861 also propose four possible criteria which may be used in designing the humanized antibodies. The first proposal was that for an acceptor, use a framework from a particular human immunoglobulin that is unusually homologous to the donor immunoglobulin to be humanized, or use a consensus framework from many human antibodies. The second proposal was that if an amino acid in the framework of the human immunoglobulin is unusual and the donor amino acid at that position is typical for human sequences, then the donor amino acid rather than the acceptor may be selected. The third proposal was that in the positions immediately adjacent to the 3 CDRs in the humanized immunoglobulin chain, the donor amino acid rather than the acceptor amino acid may be selected. The fourth proposal was to use the donor amino acid reside at the framework positions at which the amino acid is predicted to have a side chain atom within 3 A of the CDRs in a three dimensional model of the antibody and is predicted to be capable of interacting with the CDRs. The above methods are merely illustrative of some of the methods that one skilled in the art could employ to make humanized antibodies. One of ordinary skill in the art will be familiar with other methods for antibody humanization.
In one embodiment of the humanized forms of the antibodies, some, most or all of the amino acids outside the CDR regions have been replaced with amino acids from human immunoglobulin molecules but where some, most or all amino acids within one or more CDR regions are unchanged. Small additions, deletions, insertions, substitutions or modifications of amino acids are permissible as long as they would not abrogate the ability of the antibody to bind a given antigen. Suitable human immunoglobulin molecules would include IgGl, IgG2, IgG3, IgG4, IgA and IgM molecules. A "humanized" antibody retains a similar antigenic specificity as the original antibody. However, using certain methods of humanization, the affinity and/or specificity of binding of the antibody may be increased using methods of "directed evolution", as described by Wu et ah, /. Mol. Biol. 294: 151, 1999, the contents of which are incorporated herein by reference.
Fully human monoclonal antibodies also can be prepared by immunizing mice transgenic for large portions of human immunoglobulin heavy and light chain loci. See, e.g., U.S. Pat. Nos. 5,591,669, 5,598,369, 5,545,806, 5,545,807, 6,150,584, and references cited therein, the contents of which are incorporated herein by reference. These animals have been genetically modified such that there is a functional deletion in the production of endogenous (e.g., murine) antibodies. The animals are further modified to contain all or a portion of the human germ-line immunoglobulin gene locus such that immunization of these animals will result in the production of fully human antibodies to the antigen of interest. Following
immunization of these mice (e.g., XenoMouse (Abgenix), HuMAb mice (Medarex/GenPharm)), monoclonal antibodies can be prepared according to standard hybridoma technology. These monoclonal antibodies will have human immunoglobulin amino acid sequences and therefore will not provoke human anti-mouse antibody (KAMA) responses when administered to humans.
In vitro methods also exist for producing human antibodies. These include phage display technology (U.S. Pat. Nos. 5,565,332 and 5,573,905) and in vitro stimulation of human B cells (U.S. Pat. Nos. 5,229,275 and 5,567,610). The contents of these patents are incorporated herein by reference.
Thus, as will be apparent to one of ordinary skill in the art, the present invention also provides for F(ab') 2 Fab, Fv and Fd fragments; chimeric antibodies in which the Fc and/or FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric F(ab')2 fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric Fab fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; and chimeric Fd fragment antibodies in which the FR and/or CDR1 and/or CDR2 regions have been replaced by homologous human or non human sequences. The present invention also includes so-called single chain antibodies.
The various antibody molecules and fragments may derive from any of the commonly known immunoglobulin classes, including but not limited to IgA, secretory IgA, IgE, IgG and IgM. IgG subclasses are also well known to those in the art and include but are not limited to human IgGl, IgG2, IgG3 and IgG4. In a preferred embodiment, the compound of the invention is a Human IgG4.
In another embodiment, the antibody according to the invention is a single domain antibody. The term“single domain antibody” (sdAb) or "VHH" refers to the single heavy chain variable domain of antibodies of the type that can be found in Camelid mammals which are naturally devoid of light chains. Such VHH are also called“nanobody®”. According to the invention, sdAb can particularly be llama sdAb. The term“VHH” refers to the single heavy chain having 3 complementarity determining regions (CDRs): CDR1, CDR2 and CDR3. The term“complementarity determining region” or“CDR” refers to the hypervariable amino acid sequences which define the binding affinity and specificity of the VHH.
The VHH according to the invention can readily be prepared by an ordinarily skilled artisan using routine experimentation. The VHH variants and modified form thereof may be produced under any known technique in the art such as in-vitro maturation.
VHHs or sdAbs are usually generated by PCR cloning of the V-domain repertoire from blood, lymph node, or spleen cDNA obtained from immunized animals into a phage display vector, such as pHEN2. Antigen- specific VHHs are commonly selected by panning phage libraries on immobilized antigen, e.g., antigen coated onto the plastic surface of a test tube, biotinylated antigens immobilized on streptavidin beads, or membrane proteins expressed on the surface of cells. However, such VHHs often show lower affinities for their antigen than VHHs derived from animals that have received several immunizations. The high affinity of VHHs from immune libraries is attributed to the natural selection of variant VHHs during clonal expansion of B-cells in the lymphoid organs of immunized animals. The affinity of VHHs from non-immune libraries can often be improved by mimicking this strategy in vitro, i.e., by site directed mutagenesis of the CDR regions and further rounds of panning on immobilized antigen under conditions of increased stringency (higher temperature, high or low salt concentration, high or low pH, and low antigen concentrations). VHHs derived from camelid are readily expressed in and purified from the E. coli periplasm at much higher levels than the corresponding domains of conventional antibodies. VHHs generally display high solubility and stability and can also be readily produced in yeast, plant, and mammalian cells. For example, the“Hamers patents” describe methods and techniques for generating VHH against any desired target (see for example US 5,800,988; US 5,874, 541 and US 6,015,695). The“Hamers patents” more particularly describe production of VHHs in bacterial hosts such as E. coli (see for example US 6,765,087) and in lower eukaryotic hosts such as moulds (for example Aspergillus or Trichoderma) or in yeast (for example Saccharomyces, Kluyveromyces, Hansenula or Pichia) (see for example US 6,838,254).
In another aspect, the invention provides an antibody that competes for binding to the target with the antibody of the invention.
As used herein, the term "binding" in the context of the binding of an antibody to a predetermined antigen or epitope typically is a binding with an affinity corresponding to a KD of about 10-7 M or less, such as about 10-8 M or less, such as about 10-9 M or less, about 10- 10 M or less, or about 10-11 M or even less when determined by for instance surface plasmon resonance (SPR) technology in a BIAcore 3000 instrument using a soluble form of the antigen as the ligand and the antibody as the analyte. BIACORE® (GE Healthcare, Piscaataway, NJ) is one of a variety of surface plasmon resonance assay formats that are routinely used to epitope
bin panels of monoclonal antibodies. Typically, an antibody binds to the predetermined antigen with an affinity corresponding to a KD that is at least ten- fold lower, such as at least lOO-fold lower, for instance at least 1, 000-fold lower, such as at least 10,000-fold lower, for instance at least 100,000-fold lower than its KD for binding to a non-specific antigen (e.g., BSA, casein), which is not identical or closely related to the predetermined antigen. When the KD of the antibody is very low (that is, the antibody has a high affinity), then the KD with which it binds the antigen is typically at least 10,000-fold lower than its KD for a non-specific antigen. An antibody is said to essentially not bind an antigen or epitope if such binding is either not detectable (using, for example, plasmon resonance (SPR) technology in a BIAcore 3000 instrument using a soluble form of the antigen as the ligand and the antibody as the analyte), or is 100 fold, 500 fold, 1000 fold or more than 1000 fold less than the binding detected by that antibody and an antigen or epitope having a different chemical structure or amino acid sequence.
Additional antibodies can be identified based on their ability to cross-compete (e.g., to competitively inhibit the binding of, in a statistically significant manner) with other antibodies of the invention in standard antigen binding assays. The ability of a test antibody to inhibit the binding of antibodies of the present invention to the target demonstrates that the test antibody can compete with that antibody for binding to the target; such an antibody may, according to non-limiting theory, bind to the same or a related (e.g., a structurally similar or spatially proximal) epitope on the target as the antibody with which it competes. Thus, another aspect of the invention provides antibodies that bind to the same antigen as, and compete with, the antibodies disclosed herein. As used herein, an antibody“competes” for binding when the competing antibody inhibits the target binding of an antibody or antigen binding fragment of the invention by more than
50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79, 80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98 or 99% in the presence of an equimolar concentration of competing antibody.
In other embodiments the antibodies or antigen binding fragments of the invention bind to one or more epitopes of the target. In some embodiments, the epitopes to which the present antibodies or antigen binding fragments bind are linear epitopes. In other embodiments, the epitopes to which the present antibodies or antigen binding fragments bind are non-linear, conformational epitopes.
In one embodiment, the compound of the invention is an IL-10 expression inhibitor, TGFp i expression inhibitor or a FOXP3 expression inhibitor.
An“inhibitor of expression” refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene.
The target expression inhibitors for use in the present invention may be based on antisense oligonucleotide constructs. Anti-sense oligonucleotides, including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of the target mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of the target proteins, and thus activity, in a cell. For example, antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding the target can be synthesized, e.g., by conventional phosphodiester techniques and administered by e.g., intravenous injection or infusion. Methods for using antisense techniques for specifically alleviating gene expression of genes whose sequence is known are well known in the art (e.g. see U.S. Pat. Nos. 6,566,135; 6,566,131; 6,365,354; 6,410,323; 6,107,091; 6,046,321; and 5,981,732).
Small inhibitory RNAs (siRNAs) can also function as target expression inhibitors for use in the present invention. The target gene expression can be reduced by contacting the subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that the target expression is specifically inhibited (i.e. RNA interference or RNAi). Methods for selecting an appropriate dsRNA or dsRNA-encoding vector are well known in the art for genes whose sequence is known (e.g. see Tuschl, T. et al. (1999); Elbashir, S. M. et al. (2001); Hannon, GJ. (2002); McManus, MT. et al. (2002); Brummelkamp, TR. et al. (2002); U.S. Pat. Nos. 6,573,099 and 6,506,559; and International Patent Publication Nos. WO 01/36646, WO 99/32619, and WO 01/68836).
Ribozymes can also function as target expression inhibitors for use in the present invention. Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. The mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of the target mRNA sequences are thereby useful within the scope of the present invention. Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the
target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable. The suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
Both antisense oligonucleotides and ribozymes useful target inhibitors can be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules can be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences can be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention can be introduced as a means of increasing intracellular stability and half-life. Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3’ ends of the molecule, or the use of phosphorothioate or 2’-0-methyl rather than phosphodiesterase linkages within the oligonucleotide backbone.
Antisense oligonucleotides siRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector. In its broadest sense, a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide siRNA or ribozyme nucleic acid to the cells and preferably cells expressing the target. Preferably, the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector. In general, the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide siRNA or ribozyme nucleic acid sequences. Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus. One can readily employ other vectors not named but known to the art.
Preferred viral vectors are based on non-cytopathic eukaryotic viruses in which non- essential genes have been replaced with the gene of interest. Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retroviruses
have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo. Standard protocols for producing replication-deficient retroviruses (including the steps of incorporation of exogenous genetic material into a plasmid, transfection of a packaging cell lined with plasmid, production of recombinant retroviruses by the packaging cell line, collection of viral particles from tissue culture media, and infection of the target cells with viral particles) are provided in KRIEGLER (A Laboratory Manual," W.H. Freeman C.O., New York, 1990) and in MURRY ("Methods in Molecular Biology," vol.7, Humana Press, Inc., Cliffton, N.J., 1991).
Preferred viruses for certain applications are the adeno-viruses and adeno-associated viruses, which are double-stranded DNA viruses that have already been approved for human use in gene therapy. The adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions. Reportedly, the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection. In addition, wild- type adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event. The adeno-associated virus can also function in an extrachromosomal fashion.
Other vectors include plasmid vectors. Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g., SANBROOK et al, "Molecular Cloning: A Laboratory Manual," Second Edition, Cold Spring Harbor Laboratory Press, 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid. Some commonly used plasmids include pBR322, pUCl8, pUCl9, pRC/CMV, SV40, and pBlueScript. Other plasmids are well known to those of ordinary skill in the art. Additionally, plasmids may be custom designed using restriction enzymes and ligation reactions to remove and add specific fragments of DNA. Plasmids may be delivered by
a variety of parenteral, mucosal and topical routes. For example, the DNA plasmid can be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally. It may also be administered into the epidermis or a mucosal surface using a gene-gun. The plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
Typically, the inhibitors according to the invention as described above are administered to the patient in a therapeutically effective amount.
By a "therapeutically effective amount" of the inhibitor of the present invention as above described is meant a sufficient amount of the inhibitor for treating cancer at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the inhibitors and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific inhibitor employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific inhibitor employed; the duration of the treatment; drugs used in combination or coincidential with the specific inhibitor employed; and like factors well known in the medical arts. For example, it is well within the skill of the art to start doses of the inhibitor at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. However, the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day. Typically, the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the inhibitor of the present invention for the symptomatic adjustment of the dosage to the patient to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the inhibitor of the present invention, particularly from 1 mg to about 100 mg of the inhibitor of the present invention. An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
In a particular embodiment, the compound according to the invention may be used in a concentration between 0.01 mM and 20 mM, particularly, the compound of the invention may be used in a concentration of 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 20.0 mM.
According to the invention, the compound of the present invention is administered to the subject in the form of a pharmaceutical composition. Typically, the compound of the present invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions. "Pharmaceutically" or "pharmaceutically acceptable" refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate. A pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
In the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, the active principle, alone or in combination with another active principle, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings. Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
Typically, the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the
growth of microorganisms. The compound of the present invention can be formulated into a composition in a neutral or salt form. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. The carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, the composition includes isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized agent of the present inventions into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the typical methods of preparation are vacuum-drying and freeze- drying techniques which yield a powder of the compound of the present invention plus any additional desired ingredient from a previously sterile- filtered solution thereof. The preparation of more, or highly concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small tumor area. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed. For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic
with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
In some embodiments, the compound of the present invention is administered sequentially or concomitantly with one or more therapeutic active agent such as anti-cancer therapy such as immunotherapeutic agent, chemotherapeutic agent or radiotherapeutic agent.
In one embodiment, said additional active compounds may be contained in the same composition or administrated separately.
In another embodiment, the pharmaceutical composition of the invention relates to combined preparation for simultaneous, separate or sequential use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof.
The invention also provides kits comprising the compound of the invention. Kits containing the compound of the invention find use in therapeutic methods.
In a further aspect, the present invention relates to a method for treating progressive chronic lymphocytic leukemia in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound selected from the group consisting of IL-10 inhibitor, TGFp 1 inhibitor and/or FOXP3 modulator.
In a further aspect, the present invention relates to a method of treating progressive chronic lymphocytic leukemia in a patient in need thereof comprising the steps of:
(i) identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia by performing the method according to the invention, and
(ii) administering to said patient the compound of the invention and/or an anti-cancer treatment when it is concluded that the patient has or is at risk of having or developing progressive chronic lymphocytic leukemia.
In a further aspect, the present invention relates to a method of screening a candidate compound for use as a drug for treating cancer in a patient in need thereof, wherein the method comprises the steps of:
providing a IL-10, TGFp i and/or FOXP3, providing a cell, B cell, chronic lymphocytic leukemia B cells, bone marrow sample, lymphoid organ sample, blood sample, serum sample and plasma sample, tissue sample or organism expressing IL-10, TGFp i and/or FOXP3,
providing a candidate compound such as a small organic molecule, a polypeptide, an aptamer, an antibody or an intra-antibody,
quantifying the IL-10, TGFp i and/or FOXP3 expression and/or activity, and selecting positively candidate compounds that inhibit IL-10, TGFp i and/or FOXP3 expression and/or activity, and/or that modulate FOXP3 expression and/or activity.
Methods for measuring IL-10, TGFp i and/or FOXP3 activity are well known in the art (Llorente et al, 2000; Clark et al, 2013; WO 2011/143280; US2013/0109619;
WO2016196178; WO 2015/124715 ; US 2009/0325868). For example, measuring the IL-10, TGFp 1 and/or FOXP3 activity involves determining a Ki on the IL-10, TGFp 1 and/or FOXP3 cloned and transfected in a stable manner into a CHO cell line, measuring cancer cells viability/survival, measuring cancer cell migration and invasion abilities, measuring cancer cell growth, measuring cancer cell proliferation, measuring cancer cell secretion in the presence or absence of the candidate compound.
Tests and assays for screening and determining whether a candidate compound is a IL- 10, TGFp i and/or FOXP3 inhibitor, and/or FOXP3 modulator are well known in the art (Llorente et al, 2000; Clark et al, 2013; WO 2011/143280; US2013/0109619;
WO2016196178; WO 2015/124715 ; US 2009/0325868; WO2010/089443; US2011/0294734; US2012/0315256; US2007/0014767; US2013/0225655; US2010/0034786; W02013/050596). In vitro and in vivo assays may be used to assess the potency and selectivity of the candidate compounds to inhibit IL-10, TGFp 1 and/or FOXP3 activity, or to modulate FOXP3 activity.
Activities of the candidate compounds, their ability to bind IL-10, TGFp 1 and/or FOXP3 and their ability to inhibit IL-10, TGFp 1 and/or FOXP3 activity, modulate FOXP3 activity may be tested using isolated cancer cell, cancer cell lines or CHO cell line cloned and transfected in a stable manner by the human IL-10, TGFp i and/or FOXP3.
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
FIGURES:
Figure 1: CLL-B cells express immune regulatory cytokines
A-C: Analysis of purified CD5+CDl9+ CLL-B cells from blood or lymph nodes (n=3) for IL10 and TGF 1 production by flow cytometry. (A and C) Representative dot plots with the percentage of positive populations, (B) Graphic representation of ILl0+ or TGF 1 cells among blood CD5+CDl9+ cells at the time of isolation (0, n=5) or after three days of culture (3,
IL10 n=26, TGF 1 n=30) in the presence (+) or not (-) of stimulatory CD40L and anti-IgM. Dotted lines link individual patient samples. Wilcoxon signed-rank test. (C) Representative co expression of IL10 and TGF 1 . (D-E) Levels of the indicated cytokines secreted in the culture supernatant were evaluated by MSD technology (n=l7). (D right) Linear regression< between levels of TGF l in the culture supernatant and the percentage of TGF 1 cells is presented (r=0.82, n=9). Pearson’s correlation. (E) Cells were stimulated (+) or not (-) with CD40L and anti-IgM. Wilcoxon signed-rank test. *P< 0.05, ** P< 0.01; ns, not significant P >0.05.
Figure 2: CLL-B cells express FOXP3
(A-B): Analysis of purified CD5+CDl9+ CLL-B cells from blood or lymph nodes (n=3) for FOXP3 expression by flow cytometry. (A) Graphic representation of FOXP3+ cells among CD5+CDl9+ cells at the time of isolation (0, n=5) or after three days of culture (3) in the presence (+, n=34) or not (-, n=32) of CD40L and anti-IgM. Dotted lines link individual patient samples. (B). Representative co-expression of IL10 and FOXP3. The dotted line indicates that both sides are obtained from the same western blot. Wilcoxon signed-rank test, ns, not significant P >0.05.
Figure 3: Phenotypic Characterization of CLL-B cells.
(A-B) : Purified B cells were cultured for 72 hours and stained with the indicated membrane markers, and for IL-10, TGFp i or FOXP3. MFI of CD5, CD19, CD25, CD27 were compared between ILl0+/ (n=l6), TGF-b I (n= 16) and FOXP3+/ (n=l4) cells in unstimulated (A) and stimulated conditions (CD40L and anti-IgM, B). Ratio of membrane markers MFI between positive and negative cells is graphed. Wilcoxon signed-rank test * P<0.05, ** P<0.0l, ***P<0.00l,**** P<0.000l, ns, not significant P >0.05.
Figure 4: Differential expression of regulatory factors between CD5h,ghCD19h,gh and CD5d,mCD19d,m subpopulations
Purified B cells were stained for CD5 and CD 19 and analysed by flow cytometry. Cumulative results of IL10 (n=26), TGFp i (n=30) and FOXP3 (n=3 l) expression in CD5highCDl9high (H) and CD5dimCDl9dim (D) cells stimulated (+) or not (-), compared with Wilcoxon signed-rank test, * P<0.05, ** P<0.0l, ***P<0.00l,**** P<0.000l , ns, not significant P >0.05
Figure 5: Regulatory functions of CLL-B cells
(A) Purified CD4+T cells stimulated with anti-CD3 and anti-CD28 (S) or not (NS) were cultured for 48 hours in presence or not (-) with CLL-B cells activated with CD40L and anti- IgM at the indicated ratios (T/B). Scatter dot plots indicate the frequency of TNFa+ (left, n=5)
and IFNy+ (right, n=9) cells among CD4+T cells. ANOVA test *p < 0.01; ** p<0.00l . (B) Bar graphs show the frequency of FOXP3+ among CD4+ T cells and CD4+CD25+CDl27lowFOXP3+ Treg cells upon 48h co-culture (+) or not (-) with CLL-B cells (1 :1 ratio; n=5). Cells were stimulated (+) or not (-) as indicated. Wilcoxon signed-rank test * P<0.05, ** P<0.0l, ***P<0.00l . T cells stimulated (S) or not (NS) were co-cultured for 72h with CLL-B cells with the indicated ratios. Representative profile of 4 experiments is depicted.
Figure 6: A polyfunctional score is correlated with factors at risk of progression
Dimentional analysis of the polyfunctional score according to major risk factors: IGHV (Mutate, M; Unmutated, UM), ZAP70 and CD38 expressions (-, Negative and +, positive), Binet stage (A and B or C), and %MTS (Non responder, NR; Responder, R) of CLL progression in the CD5hlghCDl9hlgh subpopulation. Median and [Ql,Q3] are depicted. * P <0.05 (Mann- Whitney U test).
EXAMPLE:
Material & Methods
Patients, Materials and Methods
A cohort of 42 CLL patients was used in this study. CLL diagnosis was confirmed using international guidelines (43, 44) and approved by the local ethic committee (CLEA, GHPSSD, Avicenne hospital). The clinical and biological parameters (Service d’Hematologie Biologique de l’Hopital Avicenne) such as Binet stage at the experiment time, CD38 and ZAP-70 expressions, IGHV gene mutational status, cytogenetic features, TP53 mutation, other genetic mutations and need of treatment were analyzed (Tables 1-3).
Table 1. Patients’ biological and clinical parameters.
Table 2: CLL patients’s clinico-biological features
A. F: female and M: male
B. UM: UnMutated; M: Mutated
C. ND: not determined; POS: positive; NEG: negative
D. Metabolic activity in response to anti-IgM stimulation; R: Responder; NR: Non-Responder
Table 3: CLL features and polyfunctional score
E. 0: no treatment at experiment time; 1 : treatment at the experiment
F. 0: A at the experiment time; 1 : B or C treatment at the experiment
G. 0: Zap70 negative; 1 : Zap70 positive
H. 0: IGHV mutated; 1 : IGHV unmutated
I. 0: CD38 negative; 1 : CD38 positive
J. 0: dell7 and/or dell l negative; 1 : dell7 and/or dell l positive
K. 0: TP53 WT; 1 : TP53 mutated
L. 0: no mutated; 1 : at least one mutated gene ( NOTCH1 , POT1, RPS15, SF3B1, BIRC3, FBW7, A TM, MYD88, PLCG2 )
Human cells isolation and cell culture
B and T CD4+ lymphocytes were isolated from total blood using Rosette B and CD4+ T isolation kits (STEMCELL). Isolated B and T cells subsets purity was assessed by flow cytometry analysis and was typically > 95%. Isolated B and T cells were cultured in RPMI 1640 containing L-glutamine and supplemented with 100 U/mg/ml penicillin/streptomycin (Life Technologies, USA), and 10% ECS (Biosera) at 37°C in humidified incubator containing 5% C02 at the concentration of 2xl06/ml for 72 hours. B cells were stimulated or not with a combination of soluble CD40L (1 gg/ml; Miltenyi Biotec) and coated anti-IgM (20 gg/ml; Jackson ImmunoResearch) or a combination of soluble CD40L and IL21 (50ng/ml, Gibco by Life Technologies). CD4+ T cells were stimulated or not by coated anti-CD3 mAb (Hit-3a, 10 gg/ml; Ebioscience) and anti-CD28 mAb (CD28-2, 1 gg/ml; Ebio sciences). Lor cytokine detection by flow cytometry, Brefeldin A (BE A) (10 gg/ml; Sigma Aldrich), PMA (500 ng/ml; Sigma Aldrich) and Ionomycin (1 gg/ml; Sigma Aldrich) were added for the last 4 hours of culture. Lor B and CD4+ T cells co-culture experiments, B and T cells were mixed at different ratios (1 : 1, 1 :2, 1 :5 and 1 : 10) reaching a total cell number of 2x106 cells. MTS was assessed as previously described (5, 6).
Cell sorting
Cell sorting was performed with purified CD5+CDl9+ peripheral blood CLL-B cells further labeled with PE-Vio770 anti-CD 19 (LT19), PerCP-Vio700 anti-CD3 (REA613), APC- Vio770 anti-CD5 (UCHT2) and VioBlue anti-CD27 (M-T271) (Miltenyi Biotec). After doublet cells exclusion, gating on CDl9+, CD3 , CD5+ and CD27hlgh cells was performed. Sorted cells were then lysed to perform Western Blot. Cell-sorting experiments were performed on a LACS ARIA III cell sorter (BD Bioscience).
Western blotting
Sorted cells were lysed in NP40 lysis buffer (150 mM NaCl, 50 mM Tris-HCl, pH 7.4, 1 mM EDTA, 1% NP-40, 10% Glycerol with protease inhibitors). Proteins (2 to 20 pg) were separated on a 10% SDS-PAGE, transferred on a nitrocellulose membrane and incubated with rabbit monoclonal anti-LOXP3 antibody (D608R, Cell Signaling Technologies) and with anti- b actin mAb (AC-74, Sigma Aldrich), followed by appropriate secondary horseradish peroxidase-conjugated antibody. Detection was performed using ECL kit (Bio-Rad) and images acquired with a Chemidoc MP (BioRad).
Flow cytometry
IL10, TϋRbI and LOXP3 CLL expressing cells were first labeled for extracellular staining with anti-CD 19/V500 (HIB19), anti-CD5/V450 (UCHT2), anti-CD24/FITC (ML5),
anti-CD25/APC-Cy7 (M-A251), anti-CD27/PerCP5.5 (M-T271) and anti-CD38/PE-Cy7 (HIT2) mAbs (BD Biosciences) for 20 min. After washes, cells were fixed (2% PFA in PBS IX), permeabilized (0.5 % saponin, 1% BSA in PBS IX) and further stained with anti- IL10/APC (JES3-19F1), anti-FOXP3/PE (259D/C7) or anti-TGFp i/PE (TW4-9E7) mAbs or with their respective isotypes (BD Biosciences) overnight at 4°C.
In co-culture experiments the following alternative panel was used: anti-CD 19 A/500 (HIB19), CD5/FITC (UCHT2), CD25/APC-Cy7 (M-A251), CDl27/PE-Cy7 (SB/199) mAbs (BD Biosciences). Then, cells were fixed, permeabilized and stained with anti-IE 10/ APC (JES3-19F1), TGFp i/PE (TW4-9E7) or FOXP3/PE (259D/C7) (BD Biosciences).
For analysis of T cell division or TNFa and IFNy expression, CD4+ T cells were labeled with CellTrace Violet (Invitrogen) immediately after isolation according to the manufacter’s protocol. After co-culture for 72 hours, cells were labeled with anti-CDl9/PEVk>770 (LT19), CD3/PerCPVk>700 (REA613) and CD5/APCVio770 (UCHT2), mAbs (Miltenyi biotec) for 20 min. After washes, cells were fixed and permeabilized with cytofix/cytoperm buffer (BD biosciences) following the manufacturer’s protocol and then stained with anti-IFNy/PE (B27) (BD bioscience) and anti-TNFa/FITC (cA2) (Miltenyi biotech) or relevant isotype mAbs for 1 hour. Flow cytometry analysis was performed using a FACS Canto II driven by DIVA software (BD biosciences) and analyzed with the FlowJo software (Miltenyi Biotec).
Quantification of cytokine secretion in cell supernatants
Supernatants from B, CD4+ T and B/ CD4+ T cell cultures and co-cultures were frozen at -80°C and various cytokines (IF-l b, IF2, IF4, IF6, IF8, IF10, IF13, IF17, TNFa and IFNy, as well as TGFp 1 ) were quantified by V-plex assays (MSD) according to the manufacturer’s protocols.
Statistical analysis
Data are expressed as means with SEM or numbers with frequencies. For the comparison of qualitative values, the parametric chi-square test or Fisher's exact test was used and Mann- Whitney U test for the continuous variables or Wilcoxon signed-rank test (paired data). Association between cytokine secretion and producing cells was analyzed using Pearson correlation (r). Unsupervised hierarchical cluster analysis was performed using IF10, TGFp i and FOXP3 expression in unstimulated and stimulated state. An index called polyfunctional score was built by combining IF10, TGFp i and FOXP3 expression in each state (unstimulated or stimulated) using first component of principal components analysis. The assumption of unidimensionality of theses three factors was examined based on the eigenvalues plot (Data not
shown). All tests were two-sided at a 0.05 significance level. Analyses were carried out using R statistical software version 3.1.2 and Graphpad (Prism 7).
Results
CLL-B cells express heterogeneous immunoregulatory cytokines profile.
Purified CLL-B cells, from a cohort of 42 CLL patients displaying variable biological parameters and clinical outcome (Tables 1-2) were analyzed for their ability to express two important immunosuppressive cytokines, IL10 and TGFp i . Flow cytometry analysis of CD5+CDl9+ cells at the time of isolation (DO) and after culture for 3 days (D3) showed a highly variable proportion of cells expressing IL10 or TGFp i ranging from 0.11 to 85% and from 0.22 to 71.1%, respectively (Figure 1A-B). Comparable frequencies of ILl0+ cells were found at the time of B cell isolation and in cultured cells indicative of an IL10 constitutive expression (22.95±4.83% at DO versus 27.54±5.76% at D3) (Figure 1B). By contrast, while very low levels of TGFpl expressing cells were observed at the time of isolation; several, but not all of the cases, increased TGFp 1 expressing cells in culture indicating a high degree of inducibility (2.86±l .89% at DO versus 25.66±4.92% at D3). In order to estimate the maximum extent of inducibility, cells were stimulated by addition of CD40L and anti-IgM during the 3 days of culture followed by Phorbol Myristate Acetate (PMA), ionomycin and brefelfinA for 4 h before analysis. Upon stimulation, for most of the samples, the proportion of IL10-expressing cells (27.54±5.76% versus 29.66±6.4%) was not modified, whereas further increase of TGFp i expressing cells (25.66±4.92% versus 33.39±6.24%) was observed (Figure 1A-B). Small needle puncture analysis confirmed the presence of such ILl0+ and TGFp i subpopulations in the lymph node (Figure 1A). Examination of individual cases analyzed in all conditions (connected lines) suggested both up or downregulation only in few cases for IL10. By contrast, several cases showed further induction ofTGFp i expressing cells upon stimulation while others remained at comparable levels (Figure 1B). Nonetheless, the intracellular levels of expression of the two cytokines revealed by the median of fluorescence of the cells was not modified upon stimulation (Data not shown) Indeed, flow cytometry on CD5+CDl9+ gated cells revealed a subset of cells co-expressing IL10 and TGFp 1 (Figure 1C).
Capacity to secrete various immune modulatory cytokines was also quantified. Cells in culture for 3 days and stimulated with CD40L/anti-IgM showed low levels of IL10 (mean = 0.5 pg/ml) and high levels of TGFp i (mean = 800 pg/ml). Interestingly, TGFp i was secreted at low or high levels allowing the segregation of two groups. Furthermore, the amount of secreted TGFp i in the high secreting group was correlated to the fraction of expressing cells with levels similar to healthy controls (Figure 1D and Data not shown). All the other cytokines tested were
produced at low levels as compared to healthy controls. Similarly to TGF 1 , secretion of TNFa, IL8 and IL6 was induced upon stimulation while IL4, I L I b , IFNy , IL13, IL2 and IL17 were unchanged (Figure 1E and Data not shown). However, their production was not correlated to TGF 1 secretion (Data not shown). Granzyme B (GrB) expressing cells were rarely observed even when cells were triggered with a major inducer such as IL21/CD40L in contrast with B cells isolated from healthy subjects (Data not shown). Taken together, our results support the idea of a B-CLL cellular subpopulation, present at various extents among cases with a specific cytokine profile evocative of an immunoregulatory population that can further respond to BCR/CD40 stimulation.
CLL-B cells constitutively express FOXP3.
In line with the specific pattern of cytokines expressed, indicating similarities between regulatory and CLL-B cells, we assessed whether CLL-B cells could express, like Tregs, the major transcriptional regulator LOXP3. Llow cytometry analysis of peripheral viable CD5+CDl9+ CLL-B cells revealed a LOXP3+ subpopulation present at highly variable extents among patient samples in culture for 3 days (ranging from 0.04 to 82%; mean 20.76±3.79%) (Ligure 2A and Data not shown). Several LOXP3+ cells were also observed in small needle aspirate of lymph node and at collection of peripheral blood samples (mean 5.68±l . l9%). Stimulation for 3 days with CD40L/anti-IgM did not increase significantly the overall proportion of the LOXP3+ subpopulation (mean 21 56±4.36%) (Ligure 2A and Data not shown). Yet, a substantial up- or down-regulation of the LOXP3+ subpopulation was observed in several individual samples upon CD40L/anti-IgM triggering while others remained at comparable proportion (Ligure 2 A and Data not shown, connected lines). Mean fluorescence intensity of LOXP3 labeling did not change upon stimulation in CD5+CDl9+ patient B cells (Data not shown). Additionally, flow cytometry on CD5+CDl9+ gated cells revealed a subset of cells co expressing IL10 and LOXP3 (Ligure 2B). To confirm LOXP3 expression we performed a western blot analysis right after purification and further sorting or not using an antibody recognizing a different epitope of the transcription factor. Indeed, in CD3 CD5+CDl9+CD27+ sorted cells (Data not shown) the 47 kDa protein was expressed and enriched as compared to CD3 CD5+CDl9+ purified cells; LOXP3 was also observed in positive controls such as U20S or LOXP3 transfected HEK293T cells (Data not shown). Altogether, this analysis demonstrates that CLL-B cells display at variable extent a constitutive expression of LOXP3.
Specific subsets of CLL-B cells express IL-10, TGFpi and FOXP3 regulatory factors
Next, we determined whether a particular CLL-B subtype could account for the expression of IL-10, TGFp i and FOXP3. Purified pathological B cells were split into IL-10, TGFp i and FOXP3 positive or negative subsets and were then assessed for the level of expression (MFI) of phenotypic markers (CD5, CD 19, CD27, CD24, CD25 and CD38), previously described in various regulatory subsets (Figure 3A-B and Data not shown). As shown in Figure 3 A, three phenotypic markers e.g. CD5, CD 19 and CD27 presented significant differences of expression between IL-l0+ and IL10 cells. Comparable results were obtained between TGFpl+ and TGFpl cells. Therefore, the memory CD5+CDl9+CD27+ CLL-B cells are the IL10 and/or TGFp 1 producing subsets. We also observed that FOXP3+ cells express to a certain extent higher levels of CD5, CD19 and CD25 compared to FOXP3 cells. Analysis upon CD40/BCR triggering confirmed the results obtained with unstimulated cells in spite of a weaker CD5 detection (Figure 3B). By contrast, several other phenotypic markers usually associated to IL10 producing cells were not differentially expressed between IL-l0+/ , TGFp i or FOXP3+/ CLL-B cells and were not influenced by in vitro stimulation (Data not shown).
CD5/CD19 expression levels discriminate intra-individual heterogeneity for regulatory-markers.
In line with the specific and heterogeneous individual signature of the two phenotypic markers CD 19 and CD5 for all three regulatory factors, we distinguished for a number of samples two subpopulations of B cells at steady state: CD5hlghCDl9hlgh and CD5dimCDl9dim (Data not shown). On a set of annotated patients, both subpopulations were evaluated over a period of time. Analysis showed clearly two intra- individual profiles. Several samples exhibited remarkably homogeneous CD5 and CD 19 expression with similar CD5hlghCDl9hlgh and CD5dimCDl9dim frequencies over a long period of time (1 to two years, Group 1, Data not shown). In contrast, a second group of samples showed a pronounced heterogeneity of CD5 and CD 19 expression and a significant variation of the CD5hlghCDl9hlgh and CD5dimCDl9dim frequencies even over a shorter period of time (less than a year, Group 2, Data not shown).
Importantly, the evaluation of ILlO, TGFp i and FOXP3 expressing cells showed striking differences for every regulatory factor between the two subpopulations, both in unstimulated or CD40/BCR stimulated cells (Figure 4). This analysis defined the CD5hlghCDl9hlgh subpopulation as the major source of the regulatory factors among CLL-B cells. In comparison the CD5dimCDl9dim subset exhibited a lower ability to express IL10, TGFp i and FOXP3 (Figure 4). Altogether, these results outline the heterogeneous profile of CLL cells within a single case and a particular subset, defined as CD5hlghCDl9hlgh B cells
showing high expression levels of all three regulatory factors IL10, TGFp i and FOXP3 and diverging from already described regulatory B cells subsets.
CLL-B cells undergo a regulatory crosstalk with their T cell counterparts.
Next, we examined whether CLL-B cells might exhibit regulatory properties able to influence other immune cells. Purified autologous CD4+ T cells were cultured alone or together with CLL-B cells and their capacity to produce proinflammatory cytokines and regulatory factors was analyzed. The presence of stimulated CLL-B cells diminished, the percentage of CD4+ T cells expressing two important type 1 cytokines TNFa and IFNy. This effect was dependent on the T/ B cells ratio in the coculture (Figure 5A). Conversely, coculture between autologous T and CLL-B cells, increased the number of CD4+T cells expressing FOXP3. Since stimulation of T cells alone did not, this result argues for an orientation toward Tregs in the coculture (Figure 5B left). The modulation of FOXP3+ T cells was further supported by the assessment of CD4+CD25+CDl27lowFOXP3+ Tregs, which was significantly enhanced in the presence of CLL-B cells (Figure 5B right).
Finally, co-culture of autologous CD4+ T cells with CLL-B cells clearly impacted T cells that have a low division rate in CLL. While up to 2 division cycles were observed for anti- CD3/CD28 stimulated T cells alone during 72 h, the presence of CLL-B cells, dependent on the ratio between B and T cells in the co-culture, markedly reduced T cells division rate with full blockade for a 1 :5 ratio (Data not shown). Altogether these results indicate a positive regulatory role of CLL-B cells toward the induction of CD4+ Tregs and at contrary a suppressive role on Thl cellular response.
Expression of IL-10, TGFpi and FOXP3 in CLL-B cells correlates with disease progression.
Following the characterization of these immune regulators, we performed an unsupervised hierarchical cluster analysis of the biomarkers among the various CD5+CDl9+ patient samples. First, using the frequency of IL10, TGFp i and FOXP3 expressing cells in unstimulated and stimulated conditions, we confirmed that stimulation influenced significantly the expression of TGFpl in a number of samples. Next, the hierarchical analysis of the three parameters in both conditions showed a strong correlation between the frequency ofTGFp 1 and FOXP3 expressing cells (r=0.8) and to a lesser extent with IL10 (r=0.6). Additionally, the analysis allowed the discrimination of two clusters of samples showing, respectively, weak or elevated occurrence for the three factors. Under stimulated conditions, the variables were distributed with a higher homogeneity individualizing a cluster positive for the three parameters (Data not shown). Taking separately into account the three factors, a statistical analysis did not
reveal significant correlation with known risk factors of progression except for TGFp i that was elevated in samples positive for Zap70 (Data not shown). Next, we combined IL-10, TGFp 1 and FOXP3 expressions in a polyfunctional score, reflecting CLL-B cells capacity to express all 3 immunoregulatory factors. We challenged this score with the global or low and high CD5+CDl9+ subpopulations. The unidimensional validity of this polyfunctional score was confirmed with eigenvalues of components 2 and 3 lower than 1 in both unstimulated or stimulated conditions (Data not shown). Applying this polyfunctional score to the distinctive CD5hlghCDl9hlgh subpopulation (Figure 6) we found that values of polyfunctional score were significantly associated with IGHV status as well as with ZAP-70 positivity. These values were more elevated in IGHV unmutated or ZAP-70 positive cells. A similar analysis on CD5lowCDl9low subset or on global CLL population associated the score with Zap70 expression only.
By contrast, the novel regulatory score was never correlated with Binet staging of the disease at diagnosis, CD38 expression or metabolic activity (Figure 6 and Data not shown) (5, 6).
Remarkably, all the patients for whom the frequencies of the immonumodulating subpopulations were low (upper cluster of cases, Data not shown) experienced an indolent disease that did not require treatment. Their negative polyfunctional score was weakly associated with factors at risk of progression (20%, Table 3, UPN 14, 28, 20, 21, 24, 13). At the opposite, cases with high frequency of immuno modulating subpopulations (lower cluster of cases, Data not shown) presented with a progressive disease requiring treatment. Their positive polyfunctional score was majorly associated with factors at risk of progression (58%, Table 3, UPN 1, 23, 6, 22, 25). Moreover, a longitudinal analysis for several patients confirmed that the polyfunctional score was negative and stable for patients with an indolent disease (UPN 14, 28 and 13) while those experiencing a progression showed rise of the immunoregulatory score (UPN 2 and 8). Altogether the striking link between the capacities to detect higher levels of the three immunoregulatory factors in the tumor clone and the progressive profile of the patients argues for the validation of the polyfunctional score as a novel independent indicator of risk of progression. It also gives insights into the immunoregulatory capacity of the leukemic population.
Discussion
Recent advances in understanding the pathophysiology of CLL and the related heterogeneity at risk of progression have established the implication of dysregulated immune survey. The present study reveals important immune modulating functions of a subset of the
clonal B-CLL expansion strongly correlated to disease progression. The analysis of a cohort of 42 patients has uncovered the capacity for CD5+CDl9+CD27+ memory CLL-B cells to produce and secrete not only constitutive IL10 but also inducible TGFp 1 . An important association between the level of expression of the phenotypic markers and the heterogeneous production of the cytokines defined two subpopulations; evolution of these subpopulations is evidenced for patients encountering disease progression. Interestingly, CD5hlCDl9hl CLL-B cells also produce at variable extent the transcription factor FOXP3, a hallmark of Tregs. Co-cultures with autologous T cells have proved the functional regulatory implication of CLL-B cells in the induction of Tregs, the negative regulation of CD4+ T cell proliferation, altogether with a modulation of their pattern of secretion toward deficiency of tumor clearance. Importantly, computation of the expression of the three regulatory factors in a polyfunctional score argues for its validation as a functional predictive marker of disease progression.
Several reports have suggested implication of IL10 producing CLL-B cells in the immune deficit in CD4+ T cells observed along disease progression (20, 21, 27). The major population characterized in the initial study had phenotypic features resembling those of CD 19+CD24hlCD27+ B 10 cells, the human counterpart of murine CD 1 dhlCD5+CD 19hl B 10 cells (27). CD5+CDl9+CD24hlCD27+ cells formed a relevant subset in the CLL-B cells of our cohort (Data not shown). However, the proportions of IL-l0+, TGFp i and FOXP3+ cells in this subset were not significantly different from those found in CD24mterCD27+ or CD24+CD27 cells. In addition, both CD5+CD 19+CD24hiCD27+ and CD5+CD 19+CD24hiCD38hi subsets were able to produce IL-10 and TGFp i cytokines (Data not shown). Several other subtypes such as immature CD 19+CD24hiCD38hi, CD 1 d+CD 19+CD38+IgM+CD 147+GrB+ or CDl lb+CDl9+ cells have also been described as IL10 competent cells (18, 19, 27, 32, 34). The frequency of CD1 lb+ and CD24hlCD38hl cells among CDl9+CD5+ CLL-B cells was less than 1% and could not account for the IL-10, TGFp i and FOXP3 expressions observed (Data not shown). Also GrB+ cells did not represent a significant proportion of CLL-B cells and GrB expression was only slightly induced upon IL21/CD40L triggering as compared to healthy controls (Data not shown). Finally, mechanisms of suppression via IL10 production have also been ascribed to mature B cells that do not express typical phenotypic markers of CLL-B cells such as plasmablasts or Brl cells (18). Our results indicate that the major ILl0+ and TGFp i producing cells are memory CD5+CDl9+CD27+ that differ from the other described regulatory subtypes with markers such as CD25, CD24 and CD38. Importantly, CLL-B cells also showed differential propensity to induce expression of TGFp i , an immunoregulatory factor for Tregs induction and CD8+ T cell anergy. These two immunomodulation have also been documented
in CLL (11, 35, 36). Moreover, TGF 1 expression, when not already at the highest levels, was induced in a BCR/CD40 dependent manner, mimicking T/B cells interactions. We have previously shown that establishment of a threshold response to BCR triggering is mandatory for CLL-B cell heterogeneous survival (5). This result places the signal emanating from antigenic stimulation and crosstalk with the surrounding T cells at the center of the mechanisms responsible for a specific cytokine pattern with regulatory properties. In agreement with this central role of the BCR, we observed induction of several other cytokines involved in the remodeling of the immune response in a pathological environment such as IL6, IL8 or TNFa. The cytokines that rather promote specific helper T cells responses and pro -inflammatory signals, such as I L I b , IL2, IL4 or IFNy , were not induced and produced at very low levels as compared to healthy controls. In the pathological context, TGF 1 was induced at elevated levels while its secretion was not linked to the other cytokines. These results underline the inflection of the cytokinic balance in favor of the expansion of regulatory populations. Of note, similar ILlO+ and TGFp i subtypes were observed in lymph node aspirates from CLL patients suggesting similar modulatory mechanisms in secondary lymphoid organs.
Interestingly, CLL-B cells and mainly CD5hlghCDl9hlgh cells expressed also the transcription factor FOXP3 regardless of stimulation. Given that this factor is a major indicator of Tregs and of their immunomodulatory functions, our results demonstrated that FOXP3 is a novel indicator in CLL-B cells. A previous report described FOXP3 as present only in pre- apoptotic B cells in healthy controls (17). Therefore, we comforted our finding using different approaches including viability marker in flow cytometry and cell sorting to exclude any apoptotic cells or Tregs contamination that could account for FOXP3 expression. In CD5+CDl9+ samples we observed a strong match between the productions of the two immunoregulators FOXP3 and TGFp i . Phenotypically, FOXP3+ B cells also expressed CD25, another key marker of Tregs, which has been observed in ILlO+ Brl cells with antigen specific suppressive functions (37). Whether some coordinated regulation of FOXP3, TGFp 1 , CD5 and CD25 expressions might occur in CLL-B cell subtypes remains to be determined. A relationship between expression of CD5 and IL10 was already documented in CLL-B cells involving STAT3 and NFAT2 activation (38). Moreover, flow cytometry analysis revealed subpopulations expressing both IL10 and TGFp i or IL10 and FOXP3. Actually, FOXP3 and STAT3 that is constitutively phosphorylated in CLL are interacting partners for the expression of IL10 in a subset of regulatory T cells and a common regulation of the various proteins should be hypothesized (39).
The regulatory function of such CLL-B cells was demonstrated in co-culture experiments with autologous T cells. First, we observed a strong orientation of T cells toward a regulatory profile with the expansion of CD4+CD25+CDl27lowFOXP3+ Tregs. This orientation might result from the secretion by CLL-B cells of the two main suppressive cytokines IL10 and TGFp i (26). This expansion was accompanied by a reduction of two major CD4+ T cell populations, e.g. expressing TNF-a and IFNy targets of regulatory B cells, tumor clearance and cytotoxic response (18, 19, 40). This suppressive mechanism was dependent on the ratio in the co-culture between T and B cells and reflecting the cellular distortion observed in CLL patients. Finally, CLL-B cells hindered or suppress, in a T/B ratio dependent manner again, CD4+ T cells division, a major hallmark of Bregs. Remarkably, our analysis of the three suppressive factors expression identified the CD5hlghCDl9hlgh subpopulation as the functional subset expressing the highest levels of IL10, TGF 1 and FOXP3. This CD5hlghCDl9hlgh subset allowed distinction of two groups of cases with differential evolution during patients follow up. A reasonable interpretation of this finding is that high levels of regulatory factors provoke exhaustion of tumor survey mechanisms and tumor escape. At the opposite, the lower levels seen in CD5dimCDl9dim do not generate warning signals toward tumor survey. Among the three suppressive factors, TGF 1 only was statistically linked to ZAP70 expression or to BCR/CD40 triggering in IGHV unmutated patient samples arguing for the involvement of BCR signaling in the induction of TGFpU cells. In agreement with this result induction of TGF 1 Bregs was reported upon stimulation with CTLA-4 by T cells (30). Using T cell-independent CpG stimulation IL10 expressing cells were increased in IGHV mutated cases (21). Besides, our results indicate the presence of a constitutive pool of ILl0+ cells that was not induced upon B/T triggering.
In contrast with an individual analysis of suppressive factors, recent studies have interestingly reported a combinatorial evaluation of various cytokines or factors as better indicators of a functional subset. For instance, the balance between IL10 and TNFa expression in CDl9+CD24hlghCD38hlgh Bregs was further associated with renal allograft rejection than individual evaluation (41). Similarly, a polyfunctional index for T cells assembling expression of various cytokines was shown efficient during infectious disease response (42). Our analysis proved the benefit of using such a combinatorial score for its possible association with indicators at risk of progression of CLL patients. Computation of the three factors allowed better understanding of CLL-B cell activation and more importantly of the induction of its regulatory functions. When faced to the cohort of patient biological annotations, the polyfunctional score
of the CD5hlghCDl9hlgh subset correlated with two major indicators of B cell activation and antigenic recognition, e.g. IGHV mutational status and ZAP70 expression. Similarly, higher polyfunctional score was associated with higher number of pejorative factors and patient disease progression necessitating treatment. With a measure of three different regulatory factors, the additional information given by the polyfunctional score might therefore become a better orientation indicator of disease progression and lack of tumor clearance.
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.
1. Fabbri G, Dalla-Favera R. The molecular pathogenesis of chronic lymphocytic leukaemia. Nat Rev Cancer. 2016; 16(3): 145-62.
2. Riches JC, Ramsay AG, Gribben JG. T-cell function in chronic lymphocytic leukaemia. Semin Cancer Biol. 2010;20(6):431-8.
3. Rosenwald A, Alizadeh AA, Widhopf G, Simon R, Davis RE, Yu X, et al. Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med. 2001 ; 194(11): 1639-47.
4. Klein U, Tu Y, Stolovitzky GA, Mattioli M, Cattoretti G, Husson H, et al. Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med. 2001 ; 194(11): 1625-38.
5. Le Roy C, Deglesne PA, Chevallier N, Beitar T, Eclache V, Quettier M, et al. The degree of BCR and NFAT activation predicts clinical outcomes in chronic lymphocytic leukemia. Blood. 20l2;l20(2):356-65.
6. Deglesne PA, Chevallier N, Letestu R, Baran-Marszak F, Beitar T, Salanoubat C, et al. Survival response to B-cell receptor ligation is restricted to progressive chronic lymphocytic leukemia cells irrespective of Zap70 expression. Cancer Res. 2006;66(l4):7158- 66.
7. Muzio M, Apollonio B, Scielzo C, Frenquelli M, Vandoni I, Boussiotis V, et al. Constitutive activation of distinct BCR-signaling pathways in a subset of CLL patients: a molecular signature of anergy. Blood. 2008;l 12(1): 188-95.
8. Marklin M, Heitmann JS, Fuchs AR, Truckenmuller FM, Gutknecht M, Bugl S, et al. NFAT2 is a critical regulator of the anergic phenotype in chronic lymphocytic leukaemia. Nat Commun. 20l7;8(l):755.
9. Kipps TJ, Stevenson FK, Wu CJ, Croce CM, Packham G, Wierda WG, et al. Chronic lymphocytic leukaemia. Nat Rev Dis Primers. 20l7;3: l7008.
10. Herndon TM, Chen SS, Saba NS, Valdez J, Emson C, Gatmaitan M, et al. Direct in vivo evidence for increased proliferation of CLL cells in lymph nodes compared to bone marrow and peripheral blood. Leukemia. 2017;31 (6): 1340-7.
11. Taylor JG, Gribben JG. Microenvironment abnormalities and lymphomagenesis: Immunological aspects. Semin Cancer Biol. 2015;34:36-45.
12. Podhorecka M, Dmoszynska A, Rolinski J, Wasik E. T type l/type 2 subsets balance in B-cell chronic lymphocytic leukemia— the three-color flow cytometry analysis. Leuk Res. 2002;26(7):657-60.
13. Jitschin R, Braun M, Buttner M, Dettmer-Wilde K, Bricks J, Berger J, et al. CLL- cells induce IDOhi CDl4+HLA-DRlo myeloid-derived suppressor cells that inhibit T-cell responses and promote TRegs. Blood. 20l4;l24(5):750-60.
14. Inoue S, Mai A, Dyer MJ, Cohen GM. Inhibition of histone deacetylase class I but not class II is critical for the sensitization of leukemic cells to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. Cancer Res. 2006;66(l3):6785-92.
15. Gorosito Serran M, Fiocca Vemengo F, Beccaria CG, Acosta Rodriguez EV, Montes CL, Gruppi A. The regulatory role of B cells in autoimmunity, infections and cancer: Perspectives beyond IL10 production. FEBS Lett. 20l5;589(22):3362-9.
16. Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, and function. Immunity. 20l5;42(4):607-l2.
17. Noh J, Choi WS, Noh G, Lee JH. Presence of Foxp3 -expressing CDl9(+)CD5(+) B Cells in Human Peripheral Blood Mononuclear Cells: Human CDl9(+)CD5(+)Foxp3(+) Regulatory B Cell (Breg). Immune Netw. 20l0;l0(6):247-9.
18. Matsumoto M, Baba A, Yokota T, Nishikawa H, Ohkawa Y, Kayama H, et al. Interleukin- lO-producing plasmablasts exert regulatory function in autoimmune inflammation. Immunity. 20l4;4l(6): 1040-51.
19. Blair PA, Norena LY, Flores-Borja F, Rawlings DJ, Isenberg DA, Ehrenstein MR, et al. CDl9(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. Immunity. 20l0;32(l): l29-40.
20. DiLillo DJ, Weinberg JB, Yoshizaki A, Horikawa M, Bryant JM, Iwata Y, et al. Chronic lymphocytic leukemia and regulatory B cells share IL-10 competence and immunosuppressive function. Leukemia. 20l3;27(l): 170-82.
21. Drennan S, D'Avola A, Gao Y, Weigel C, Chrysostomou E, Steele AJ, et al. IL- 10 production by CLL cells is enhanced in the anergic IGHV mutated subset and associates with reduced DNA methylation of the IL10 locus. Leukemia. 2017.
22. Baumgarth N. The double life of a B-l cell: self-reactivity selects for protective effector functions. Nat Rev Immunol. 201 l;l l(l):34-46.
23. Horikawa M, Minard-Colin V, Matsushita T, Tedder TF. Regulatory B cell production of IL-10 inhibits lymphoma depletion during CD20 immunotherapy in mice. J Clin Invest. 2011 ;121(11):4268-80.
24. Carter NA, Vasconcellos R, Rosser EC, Tulone C, Munoz-Suano A, Kamanaka M, et al. Mice lacking endogenous IL-lO-producing regulatory B cells develop exacerbated disease and present with an increased frequency of Thl/Thl7 but a decrease in regulatory T cells. J Immunol. 2011 ; 186(10):5569-79.
25. Flores-Borja F, Bosma A, Ng D, Reddy V, Ehrenstein MR, Isenberg DA, et al. CDl9+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation. Sci Transl Med. 20l3;5(l73): l73ra23.
26. Mauri C, Menon M. The expanding family of regulatory B cells. Int Immunol. 20l5;27(l0):479-86.
27. Iwata Y, Matsushita T, Horikawa M, Dilillo DJ, Yanaba K, Venturi GM, et al. Characterization of a rare IL-lO-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood. 2011 ; 1 17 (2) : 530-41.
28. Shalapour S, Font-Burgada J, Di Caro G, Zhong Z, Sanchez-Lopez E, Dhar D, et al. Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature. 20l5;52l(7550):94-8.
29. Hagn M, Blackwell SE, Beyer T, Ebel V, Fabricius D, Lindner S, et al. B-CLL cells acquire APC- and CTL-like phenotypic characteristics after stimulation with CpG ODN and IL-21. Int Immunol. 20l4;26(7):383-95.
30. Nouel A, Pochard P, Simon Q, Segalen I, Le Meur Y, Pers JO, et al. B-Cells induce regulatory T cells through TGF-b/IϋO production in A CTLA-4 dependent manner. J Autoimmun. 2015;59:53-60.
31. Hamblin TJ, Oscier DG. Chronic lymphocytic leukaemia: the nature of the leukaemic cell. Blood Rev. l997;l 1(3): 119-28.
32. Griffin DO, Holodick NE, Rothstein TL. Human Bl cells in umbilical cord and adult peripheral blood express the novel phenotype CD20+ CD27+ CD43+ CD70-. J Exp Med. 2011 ;208(l):67-80.
33. Chiorazzi N, Ferrarini M. B cell chronic lymphocytic leukemia: lessons learned from studies of the B cell antigen receptor. Annu Rev Immunol. 2003 ;21 : 841-94.
34. Lindner S, Dahlke K, Sontheimer K, Hagn M, Kaltenmeier C, Barth TF, et al. Interleukin 21 -induced granzyme B-expressing B cells infiltrate tumors and regulate T cells. Cancer Res. 20l3;73(8):2468-79.
35. Parekh VV, Prasad DV, Banerjee PP, Joshi BN, Kumar A, Mishra GC. B cells activated by lipopolysaccharide, but not by anti-Ig and anti-CD40 antibody, induce anergy in CD8+ T cells: role of TGF-beta 1. J Immunol. 2003; 170(12):5897-911.
36. Jak M, Mous R, Remmerswaal EB, Spijker R, Jaspers A, Yague A, et al. Enhanced formation and survival of CD4+ CD25hi Foxp3+ T-cells in chronic lymphocytic leukemia. Leuk Lymphoma. 2009;50(5):788-80l .
37. Vadasz Z, Peri R, Eiza N, Slobodin G, Balbir-Gurman A, Toubi E. The Expansion of CD25 high IL-10 high FoxP3 high B Regulatory Cells Is in Association with SLE Disease Activity. J Immunol Res. 20l5;20l5:254245.
38. Garaud S, Morva A, Lemoine S, Hillion S, Bordron A, Pers JO, et al. CD5 promotes IL-10 production in chronic lymphocytic leukemia B cells through STAT3 and NFAT2 activation. J Immunol. 2011 ; 186(8):4835-44.
39. Hossain DM, Panda AK, Manna A, Mohanty S, Bhattachaqee P, Bhattacharyya S, et al. FoxP3 acts as a cotranscription factor with STAT3 in tumor-induced regulatory T cells. Immunity. 20l3;39(6): 1057-69.
40. Lemoine S, Morva A, Youinou P, Jamin C. Human T cells induce their own regulation through activation of B cells. J Autoimmun. 2011;36(3-4):228-38.
41. Cherukuri A, Rothstein DM, Clark B, Carter CR, Davison A, Hemandez- Fuentes M, et al. Immunologic human renal allograft injury associates with an altered IL- lO/TNF-a expression ratio in regulatory B cells. J Am Soc Nephrol. 20l4;25(7): 1575-85.
42. Larsen M, Arnaud L, Hie M, Parizot C, Dorgham K, Shoukry M, et al. Multiparameter grouping delineates heterogeneous populations of human IL-17 and/or IL-22 T-cell producers that share antigen specificities with other T-cell subsets. Eur J Immunol. 201 l;4l(9):2596-605.
43. Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Dohner H, et al. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood. 2008;l 1 l(l2):5446-56.
44. Eichhorst B, Robak T, Montserrat E, Ghia P, Hillmen P, Hallek M, et al. Chronic lymphocytic leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 20l5;26 Suppl 5:v78-84.
45. Llorente, et al., Clinical and biologic effects of antiinterleukin- 10 monoclonal antibody administration in systemic lupus erythematosus, Arthritis Rheum. 43 (8) (2000) 1790-
1800.
46. Clark DN, Markham JL, Sloan CS, Poole BD. Cytokine inhibition as a strategy for treating systemic lupus erythematosus. Clin Immunol. 2013 Sep;l48(3):335-43.
47. Casares N, Rudilla F, Arribillaga L, Llopiz D, Riezu-Boj JI, Lozano T, Lopez- Sagaseta J, Guembe L, Sarobe P, Prieto J, Borras-Cuesta F, Lasarte JJ. A peptide inhibitor of
FOXP3 impairs regulatory T cell activity and improves vaccine efficacy in mice. J Immunol. 2010 Nov 1 ; 185(9):5150-9.
48. Teresa Lozano, Marta Gorraiz, Aritz Lasarte-Cia, Marta Ruiz, Obdulia Rabal, Julen Oyarzabal, Sandra Hervas-Stubbs, Diana Llopiz, Pablo Sarobe, Jesus Prieto, Noelia Casares, and Juan Jose Lasarte. Blockage of FOXP3 transcription factor dimerization and FOXP3/AML1 interaction inhibits T regulatory cell activity: sequence optimization of a peptide inhibitor. Oncotarget. 2017 Sep 22; 8(42): 71709-71724.
Claims
1. A method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
i) determining the level of IL-10, TGFp i and FOXP3 or the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 in a biological sample obtained from the patient,
ii) comparing the levels determined at step i) with a reference value, and
iii) wherein detecting differential in the level of IL-10, TGFp i and FOXP3 or the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 between the biological sample and the reference value is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia.
2. A method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
i) determining the level of IL-10, TGFp i and FOXP3 or the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 in a biological sample obtained from the patient at time tl,
ii) determining the level of IL-10, TGFp i and FOXP3 or the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 in a biological sample obtained from the patient at time t2, and
iii) an increase of the levels determined at step i) and ii) between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
3. A method of identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia comprising the steps of:
i) determining the level of IL-10, TGFp i and FOXP3 or the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 in a biological sample obtained from the patient at time tl,
ii) determining the level of IL-10, TGFp i and FOXP3 or the level of cells expressing IL-10, cells expressing TGFp 1 , and cells expressing FOXP3 in a biological sample obtained from the patient at time t2,
iii) building a polyfunctional score combining the levels determined at step i) and a polyfunctional score combining the levels determined at step ii), and
iv) an increase of the polyfunctional score between tl and t2 is indicative of patient having or at risk of having or developing progressive chronic lymphocytic leukemia or resisting to treatment.
4. A compound selected from the group consisting of IL-10 inhibitor, TGFp i inhibitor and/or FOXP3 modulator for use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof.
5. A compound selected from the group consisting of IL-10 inhibitor, TGFp i inhibitor and/or FOXP3 modulator for use in the treatment of progressive chronic lymphocytic leukemia in a patient in need thereof wherein the patient was being classified as having or at risk of having or developing progressive chronic lymphocytic leukemia by the method according to any of claims 1 to 3.
6. A method for treating progressive chronic lymphocytic leukemia in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound selected from the group consisting of IL-10 inhibitor, TGFp i inhibitor and/or FOXP3 modulator.
7. A method of treating progressive chronic lymphocytic leukemia in a patient in need thereof comprising the steps of:
(i) identifying a patient having or at risk of having or developing progressive chronic lymphocytic leukemia by performing the method according to any of claims 1 to 3, and
(ii) administering to said patient the compound according to claim 4 and/or an anti-cancer treatment when it is concluded that the patient has or is at risk of having or developing progressive chronic lymphocytic leukemia.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP18305700.9 | 2018-06-08 | ||
EP18305700 | 2018-06-08 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2019234221A1 true WO2019234221A1 (en) | 2019-12-12 |
Family
ID=62748885
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2019/064946 WO2019234221A1 (en) | 2018-06-08 | 2019-06-07 | Methods for stratification and treatment of a patient suffering from chronic lymphocytic leukemia |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2019234221A1 (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113252900A (en) * | 2021-06-19 | 2021-08-13 | 山东第一医科大学附属省立医院(山东省立医院) | ApoA-based chronic lymphocytic leukemia prognosis risk assessment system and application thereof |
CN115261469A (en) * | 2022-06-23 | 2022-11-01 | 山东第一医科大学附属省立医院(山东省立医院) | Application of BRD9 in the diagnosis and treatment of chronic lymphocytic leukemia |
Citations (70)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP0126450A2 (en) | 1983-05-19 | 1984-11-28 | Ioannis Dr. Tripatzis | Particle and method for the detection of antigens and/or antibodies using this particle |
US4499052A (en) | 1982-08-30 | 1985-02-12 | Becton, Dickinson And Company | Apparatus for distinguishing multiple subpopulations of cells |
US4683202A (en) | 1985-03-28 | 1987-07-28 | Cetus Corporation | Process for amplifying nucleic acid sequences |
US4717655A (en) | 1982-08-30 | 1988-01-05 | Becton, Dickinson And Company | Method and apparatus for distinguishing multiple subpopulations of cells |
US4816567A (en) | 1983-04-08 | 1989-03-28 | Genentech, Inc. | Recombinant immunoglobin preparations |
WO1990007861A1 (en) | 1988-12-28 | 1990-07-26 | Protein Design Labs, Inc. | CHIMERIC IMMUNOGLOBULINS SPECIFIC FOR p55 TAC PROTEIN OF THE IL-2 RECEPTOR |
US5225539A (en) | 1986-03-27 | 1993-07-06 | Medical Research Council | Recombinant altered antibodies and methods of making altered antibodies |
US5229275A (en) | 1990-04-26 | 1993-07-20 | Akzo N.V. | In-vitro method for producing antigen-specific human monoclonal antibodies |
US5545807A (en) | 1988-10-12 | 1996-08-13 | The Babraham Institute | Production of antibodies from transgenic animals |
US5545806A (en) | 1990-08-29 | 1996-08-13 | Genpharm International, Inc. | Ransgenic non-human animals for producing heterologous antibodies |
US5565332A (en) | 1991-09-23 | 1996-10-15 | Medical Research Council | Production of chimeric antibodies - a combinatorial approach |
US5567610A (en) | 1986-09-04 | 1996-10-22 | Bioinvent International Ab | Method of producing human monoclonal antibodies and kit therefor |
US5573905A (en) | 1992-03-30 | 1996-11-12 | The Scripps Research Institute | Encoded combinatorial chemical libraries |
US5585089A (en) | 1988-12-28 | 1996-12-17 | Protein Design Labs, Inc. | Humanized immunoglobulins |
US5591669A (en) | 1988-12-05 | 1997-01-07 | Genpharm International, Inc. | Transgenic mice depleted in a mature lymphocytic cell-type |
US5598369A (en) | 1994-06-28 | 1997-01-28 | Advanced Micro Devices, Inc. | Flash EEPROM array with floating substrate erase operation |
US5618829A (en) | 1993-01-28 | 1997-04-08 | Mitsubishi Chemical Corporation | Tyrosine kinase inhibitors and benzoylacrylamide derivatives |
US5639757A (en) | 1995-05-23 | 1997-06-17 | Pfizer Inc. | 4-aminopyrrolo[2,3-d]pyrimidines as tyrosine kinase inhibitors |
US5728868A (en) | 1993-07-15 | 1998-03-17 | Cancer Research Campaign Technology Limited | Prodrugs of protein tyrosine kinase inhibitors |
US5800988A (en) | 1992-08-21 | 1998-09-01 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US5804396A (en) | 1994-10-12 | 1998-09-08 | Sugen, Inc. | Assay for agents active in proliferative disorders |
US5854033A (en) | 1995-11-21 | 1998-12-29 | Yale University | Rolling circle replication reporter systems |
US5859205A (en) | 1989-12-21 | 1999-01-12 | Celltech Limited | Humanised antibodies |
WO1999032619A1 (en) | 1997-12-23 | 1999-07-01 | The Carnegie Institution Of Washington | Genetic inhibition by double-stranded rna |
US5981732A (en) | 1998-12-04 | 1999-11-09 | Isis Pharmaceuticals Inc. | Antisense modulation of G-alpha-13 expression |
US6046321A (en) | 1999-04-09 | 2000-04-04 | Isis Pharmaceuticals Inc. | Antisense modulation of G-alpha-i1 expression |
US6100254A (en) | 1997-10-10 | 2000-08-08 | Board Of Regents, The University Of Texas System | Inhibitors of protein tyrosine kinases |
US6107091A (en) | 1998-12-03 | 2000-08-22 | Isis Pharmaceuticals Inc. | Antisense inhibition of G-alpha-16 expression |
US6127374A (en) | 1997-07-29 | 2000-10-03 | Warner-Lambert Company | Irreversible inhibitors of tyrosine kinases |
US6150584A (en) | 1990-01-12 | 2000-11-21 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
WO2001036646A1 (en) | 1999-11-19 | 2001-05-25 | Cancer Research Ventures Limited | Inhibiting gene expression with dsrna |
US6245759B1 (en) | 1999-03-11 | 2001-06-12 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
WO2001068836A2 (en) | 2000-03-16 | 2001-09-20 | Genetica, Inc. | Methods and compositions for rna interference |
US6306874B1 (en) | 1999-10-19 | 2001-10-23 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6313138B1 (en) | 2000-02-25 | 2001-11-06 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6316444B1 (en) | 1999-06-30 | 2001-11-13 | Merck & Co., Inc. | SRC kinase inhibitor compounds |
US6329380B1 (en) | 1999-06-30 | 2001-12-11 | Merck & Co., Inc. | SRC kinase inhibitor compounds |
US6344459B1 (en) | 1996-04-12 | 2002-02-05 | Warner-Lambert Company | Irreversible inhibitors of tyrosine kinases |
US6365354B1 (en) | 2000-07-31 | 2002-04-02 | Isis Pharmaceuticals, Inc. | Antisense modulation of lysophospholipase I expression |
US6410323B1 (en) | 1999-08-31 | 2002-06-25 | Isis Pharmaceuticals, Inc. | Antisense modulation of human Rho family gene expression |
US6420382B2 (en) | 2000-02-25 | 2002-07-16 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6479512B1 (en) | 1999-10-19 | 2002-11-12 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6498165B1 (en) | 1999-06-30 | 2002-12-24 | Merck & Co., Inc. | Src kinase inhibitor compounds |
US6566131B1 (en) | 2000-10-04 | 2003-05-20 | Isis Pharmaceuticals, Inc. | Antisense modulation of Smad6 expression |
US6566135B1 (en) | 2000-10-04 | 2003-05-20 | Isis Pharmaceuticals, Inc. | Antisense modulation of caspase 6 expression |
US6573099B2 (en) | 1998-03-20 | 2003-06-03 | Benitec Australia, Ltd. | Genetic constructs for delaying or repressing the expression of a target gene |
US6586423B2 (en) | 1999-09-10 | 2003-07-01 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6740665B1 (en) | 1999-02-10 | 2004-05-25 | Ramachandran Murali | Tyrosine kinase inhibitors and methods of using the same |
US6765087B1 (en) | 1992-08-21 | 2004-07-20 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US6794393B1 (en) | 1999-10-19 | 2004-09-21 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6838254B1 (en) | 1993-04-29 | 2005-01-04 | Conopco, Inc. | Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae |
US6875767B2 (en) | 2001-06-22 | 2005-04-05 | Merck & Co., Inc. | (5-cyano-2-thiazolyl)amino-4-pyridine tyrosine kinase inhibitors |
US6927293B2 (en) | 2001-08-30 | 2005-08-09 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6958340B2 (en) | 2001-08-01 | 2005-10-25 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US7109304B2 (en) | 2003-07-31 | 2006-09-19 | Immunomedics, Inc. | Humanized anti-CD19 antibodies |
US20070014767A1 (en) | 1998-11-24 | 2007-01-18 | Ezquerro Saenz Ignacio J | TGFbeta1-inhibitor peptides |
WO2007011856A2 (en) * | 2005-07-15 | 2007-01-25 | The Trustees Of Columbia University In The City Of New York | Compositions and methods for differential diagnosis of chronic lymphocytic leukemia |
EP1777523A1 (en) * | 2005-10-19 | 2007-04-25 | INSERM (Institut National de la Santé et de la Recherche Médicale) | An in vitro method for the prognosis of progression of a cancer and of the outcome in a patient and means for performing said method |
US20070254295A1 (en) | 2006-03-17 | 2007-11-01 | Prometheus Laboratories Inc. | Methods of predicting and monitoring tyrosine kinase inhibitor therapy |
WO2008141282A2 (en) * | 2007-05-11 | 2008-11-20 | The Regents Of The University Of Michigan | Materials and methods for foxp3 tumor suppression |
US20100034786A1 (en) | 2006-12-14 | 2010-02-11 | Medical Research Council | Use of PI3K M-TOR and Akt Inhibitors to Induce FOXP3 Expression and Generate Regulatory T Cells |
WO2010089443A2 (en) | 2009-02-05 | 2010-08-12 | Digna Biotech,S.L. | PHARMACEUTICAL COMPOSITIONS COMPRISING TGF- β1 INHIBITOR PEPTIDES |
WO2011143280A2 (en) | 2010-05-11 | 2011-11-17 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | Peptide-based inhibitor of interleukin-10 or interferon-gamma signaling |
US20120315256A1 (en) | 2010-02-22 | 2012-12-13 | Digna Biotech, S.I. | Use of transforming growth factor - beta 1 (tgf-b1) inhibitor peptides for the treatment of corneal fibrosis and/or haze |
WO2013050596A1 (en) | 2011-10-06 | 2013-04-11 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Method of maintaining foxp3 expression in expanded t regulatory cell |
US20130225655A1 (en) | 2010-05-04 | 2013-08-29 | Patrick Y. Lu | Combinations of TGFBeta and COX-2 Inhibitors and Methods for Their Therapeutic Application |
WO2015124715A1 (en) | 2014-02-21 | 2015-08-27 | Cellectis | Method for in situ inhibition of regulatory t cells |
WO2016196178A1 (en) | 2015-05-29 | 2016-12-08 | Merck Sharp & Dohme Corp. | Combination of an anti-il-10 antibody and a cpg-c type oligonucleotide for treating cancer |
WO2017067681A1 (en) * | 2015-10-19 | 2017-04-27 | Flexopharm Brain Gmbh & Co. Kg | Supporting immunomodulatory agent |
WO2017109774A1 (en) * | 2015-12-20 | 2017-06-29 | The National Institute for Biotechnology in the Negev Ltd. | Biomarkers of chronic lymphocytic leukemia and use thereof |
-
2019
- 2019-06-07 WO PCT/EP2019/064946 patent/WO2019234221A1/en active Application Filing
Patent Citations (82)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4499052A (en) | 1982-08-30 | 1985-02-12 | Becton, Dickinson And Company | Apparatus for distinguishing multiple subpopulations of cells |
US4717655A (en) | 1982-08-30 | 1988-01-05 | Becton, Dickinson And Company | Method and apparatus for distinguishing multiple subpopulations of cells |
US4816567A (en) | 1983-04-08 | 1989-03-28 | Genentech, Inc. | Recombinant immunoglobin preparations |
EP0126450A2 (en) | 1983-05-19 | 1984-11-28 | Ioannis Dr. Tripatzis | Particle and method for the detection of antigens and/or antibodies using this particle |
US4683202A (en) | 1985-03-28 | 1987-07-28 | Cetus Corporation | Process for amplifying nucleic acid sequences |
US4683202B1 (en) | 1985-03-28 | 1990-11-27 | Cetus Corp | |
US5225539A (en) | 1986-03-27 | 1993-07-06 | Medical Research Council | Recombinant altered antibodies and methods of making altered antibodies |
US5567610A (en) | 1986-09-04 | 1996-10-22 | Bioinvent International Ab | Method of producing human monoclonal antibodies and kit therefor |
US5545807A (en) | 1988-10-12 | 1996-08-13 | The Babraham Institute | Production of antibodies from transgenic animals |
US5591669A (en) | 1988-12-05 | 1997-01-07 | Genpharm International, Inc. | Transgenic mice depleted in a mature lymphocytic cell-type |
WO1990007861A1 (en) | 1988-12-28 | 1990-07-26 | Protein Design Labs, Inc. | CHIMERIC IMMUNOGLOBULINS SPECIFIC FOR p55 TAC PROTEIN OF THE IL-2 RECEPTOR |
US5585089A (en) | 1988-12-28 | 1996-12-17 | Protein Design Labs, Inc. | Humanized immunoglobulins |
US5693761A (en) | 1988-12-28 | 1997-12-02 | Protein Design Labs, Inc. | Polynucleotides encoding improved humanized immunoglobulins |
US5693762A (en) | 1988-12-28 | 1997-12-02 | Protein Design Labs, Inc. | Humanized immunoglobulins |
US5859205A (en) | 1989-12-21 | 1999-01-12 | Celltech Limited | Humanised antibodies |
US6150584A (en) | 1990-01-12 | 2000-11-21 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
US5229275A (en) | 1990-04-26 | 1993-07-20 | Akzo N.V. | In-vitro method for producing antigen-specific human monoclonal antibodies |
US5545806A (en) | 1990-08-29 | 1996-08-13 | Genpharm International, Inc. | Ransgenic non-human animals for producing heterologous antibodies |
US5565332A (en) | 1991-09-23 | 1996-10-15 | Medical Research Council | Production of chimeric antibodies - a combinatorial approach |
US5573905A (en) | 1992-03-30 | 1996-11-12 | The Scripps Research Institute | Encoded combinatorial chemical libraries |
US5800988A (en) | 1992-08-21 | 1998-09-01 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US6765087B1 (en) | 1992-08-21 | 2004-07-20 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US6015695A (en) | 1992-08-21 | 2000-01-18 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US5874541A (en) | 1992-08-21 | 1999-02-23 | Vrije Universiteit | Immunoglobulins devoid of light chains |
US5618829A (en) | 1993-01-28 | 1997-04-08 | Mitsubishi Chemical Corporation | Tyrosine kinase inhibitors and benzoylacrylamide derivatives |
US6838254B1 (en) | 1993-04-29 | 2005-01-04 | Conopco, Inc. | Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae |
US5728868A (en) | 1993-07-15 | 1998-03-17 | Cancer Research Campaign Technology Limited | Prodrugs of protein tyrosine kinase inhibitors |
US5598369A (en) | 1994-06-28 | 1997-01-28 | Advanced Micro Devices, Inc. | Flash EEPROM array with floating substrate erase operation |
US5804396A (en) | 1994-10-12 | 1998-09-08 | Sugen, Inc. | Assay for agents active in proliferative disorders |
US5639757A (en) | 1995-05-23 | 1997-06-17 | Pfizer Inc. | 4-aminopyrrolo[2,3-d]pyrimidines as tyrosine kinase inhibitors |
US5854033A (en) | 1995-11-21 | 1998-12-29 | Yale University | Rolling circle replication reporter systems |
US6344459B1 (en) | 1996-04-12 | 2002-02-05 | Warner-Lambert Company | Irreversible inhibitors of tyrosine kinases |
US6127374A (en) | 1997-07-29 | 2000-10-03 | Warner-Lambert Company | Irreversible inhibitors of tyrosine kinases |
US6562818B1 (en) | 1997-07-29 | 2003-05-13 | Warner-Lambert Company | Irreversible inhibitors of tyrosine kinases |
US6100254A (en) | 1997-10-10 | 2000-08-08 | Board Of Regents, The University Of Texas System | Inhibitors of protein tyrosine kinases |
WO1999032619A1 (en) | 1997-12-23 | 1999-07-01 | The Carnegie Institution Of Washington | Genetic inhibition by double-stranded rna |
US6506559B1 (en) | 1997-12-23 | 2003-01-14 | Carnegie Institute Of Washington | Genetic inhibition by double-stranded RNA |
US6573099B2 (en) | 1998-03-20 | 2003-06-03 | Benitec Australia, Ltd. | Genetic constructs for delaying or repressing the expression of a target gene |
US20070014767A1 (en) | 1998-11-24 | 2007-01-18 | Ezquerro Saenz Ignacio J | TGFbeta1-inhibitor peptides |
US6107091A (en) | 1998-12-03 | 2000-08-22 | Isis Pharmaceuticals Inc. | Antisense inhibition of G-alpha-16 expression |
US5981732A (en) | 1998-12-04 | 1999-11-09 | Isis Pharmaceuticals Inc. | Antisense modulation of G-alpha-13 expression |
US6740665B1 (en) | 1999-02-10 | 2004-05-25 | Ramachandran Murali | Tyrosine kinase inhibitors and methods of using the same |
US6245759B1 (en) | 1999-03-11 | 2001-06-12 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6544988B1 (en) | 1999-03-11 | 2003-04-08 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6046321A (en) | 1999-04-09 | 2000-04-04 | Isis Pharmaceuticals Inc. | Antisense modulation of G-alpha-i1 expression |
US6329380B1 (en) | 1999-06-30 | 2001-12-11 | Merck & Co., Inc. | SRC kinase inhibitor compounds |
US6498165B1 (en) | 1999-06-30 | 2002-12-24 | Merck & Co., Inc. | Src kinase inhibitor compounds |
US6316444B1 (en) | 1999-06-30 | 2001-11-13 | Merck & Co., Inc. | SRC kinase inhibitor compounds |
US6410323B1 (en) | 1999-08-31 | 2002-06-25 | Isis Pharmaceuticals, Inc. | Antisense modulation of human Rho family gene expression |
US6586423B2 (en) | 1999-09-10 | 2003-07-01 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6586424B2 (en) | 1999-09-10 | 2003-07-01 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6479512B1 (en) | 1999-10-19 | 2002-11-12 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6794393B1 (en) | 1999-10-19 | 2004-09-21 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6306874B1 (en) | 1999-10-19 | 2001-10-23 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
WO2001036646A1 (en) | 1999-11-19 | 2001-05-25 | Cancer Research Ventures Limited | Inhibiting gene expression with dsrna |
US6313138B1 (en) | 2000-02-25 | 2001-11-06 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6420382B2 (en) | 2000-02-25 | 2002-07-16 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
WO2001068836A2 (en) | 2000-03-16 | 2001-09-20 | Genetica, Inc. | Methods and compositions for rna interference |
US6365354B1 (en) | 2000-07-31 | 2002-04-02 | Isis Pharmaceuticals, Inc. | Antisense modulation of lysophospholipase I expression |
US6566135B1 (en) | 2000-10-04 | 2003-05-20 | Isis Pharmaceuticals, Inc. | Antisense modulation of caspase 6 expression |
US6566131B1 (en) | 2000-10-04 | 2003-05-20 | Isis Pharmaceuticals, Inc. | Antisense modulation of Smad6 expression |
US6875767B2 (en) | 2001-06-22 | 2005-04-05 | Merck & Co., Inc. | (5-cyano-2-thiazolyl)amino-4-pyridine tyrosine kinase inhibitors |
US6958340B2 (en) | 2001-08-01 | 2005-10-25 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US6927293B2 (en) | 2001-08-30 | 2005-08-09 | Merck & Co., Inc. | Tyrosine kinase inhibitors |
US7109304B2 (en) | 2003-07-31 | 2006-09-19 | Immunomedics, Inc. | Humanized anti-CD19 antibodies |
WO2007011856A2 (en) * | 2005-07-15 | 2007-01-25 | The Trustees Of Columbia University In The City Of New York | Compositions and methods for differential diagnosis of chronic lymphocytic leukemia |
EP1777523A1 (en) * | 2005-10-19 | 2007-04-25 | INSERM (Institut National de la Santé et de la Recherche Médicale) | An in vitro method for the prognosis of progression of a cancer and of the outcome in a patient and means for performing said method |
US20070254295A1 (en) | 2006-03-17 | 2007-11-01 | Prometheus Laboratories Inc. | Methods of predicting and monitoring tyrosine kinase inhibitor therapy |
US20100034786A1 (en) | 2006-12-14 | 2010-02-11 | Medical Research Council | Use of PI3K M-TOR and Akt Inhibitors to Induce FOXP3 Expression and Generate Regulatory T Cells |
US20090325868A1 (en) | 2007-05-11 | 2009-12-31 | The Regents Of The University Of Michigan | Materials And Methods For FOXP3 Tumor Suppression |
WO2008141282A2 (en) * | 2007-05-11 | 2008-11-20 | The Regents Of The University Of Michigan | Materials and methods for foxp3 tumor suppression |
US20110294734A1 (en) | 2009-02-05 | 2011-12-01 | Digna Biotech, S.L. | Pharmaceutical compositions comprising tgf-beta 1 inhibitor peptides |
WO2010089443A2 (en) | 2009-02-05 | 2010-08-12 | Digna Biotech,S.L. | PHARMACEUTICAL COMPOSITIONS COMPRISING TGF- β1 INHIBITOR PEPTIDES |
US20120315256A1 (en) | 2010-02-22 | 2012-12-13 | Digna Biotech, S.I. | Use of transforming growth factor - beta 1 (tgf-b1) inhibitor peptides for the treatment of corneal fibrosis and/or haze |
US20130225655A1 (en) | 2010-05-04 | 2013-08-29 | Patrick Y. Lu | Combinations of TGFBeta and COX-2 Inhibitors and Methods for Their Therapeutic Application |
WO2011143280A2 (en) | 2010-05-11 | 2011-11-17 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | Peptide-based inhibitor of interleukin-10 or interferon-gamma signaling |
US20130109619A1 (en) | 2010-05-11 | 2013-05-02 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Serv | Peptide-based inhibitor of interleukin-10 or interferon-gamma signaling |
WO2013050596A1 (en) | 2011-10-06 | 2013-04-11 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Method of maintaining foxp3 expression in expanded t regulatory cell |
WO2015124715A1 (en) | 2014-02-21 | 2015-08-27 | Cellectis | Method for in situ inhibition of regulatory t cells |
WO2016196178A1 (en) | 2015-05-29 | 2016-12-08 | Merck Sharp & Dohme Corp. | Combination of an anti-il-10 antibody and a cpg-c type oligonucleotide for treating cancer |
WO2017067681A1 (en) * | 2015-10-19 | 2017-04-27 | Flexopharm Brain Gmbh & Co. Kg | Supporting immunomodulatory agent |
WO2017109774A1 (en) * | 2015-12-20 | 2017-06-29 | The National Institute for Biotechnology in the Negev Ltd. | Biomarkers of chronic lymphocytic leukemia and use thereof |
Non-Patent Citations (62)
Title |
---|
"Biochemistry and Immunology", 1996, ACADEMIC PRESS, article "Coding, Monoclonal Antibodies: Principles and Practice: Production and Application of Monoclonal Antibodies in Cell Biology" |
AGNEW CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186 |
BAUMGARTH N: "The double life of a B-1 cell: self-reactivity selects for protective effector functions", NAT REV IMMUNOL, vol. 1 1, no. 1, 2011, pages 34 - 46 |
BEYER M. ET AL: "Reduced frequencies and suppressive function of CD4+CD25hi regulatory T cells in patients with chronic lymphocytic leukemia after therapy with fludarabine.", BLOOD, vol. 106, no. 6, 15 September 2005 (2005-09-15), pages 2018 - 2025, XP009083288 * |
BLAIR PANORENA LYFLORES-BORJA FRAWLINGS DJISENBERG DAEHRENSTEIN MR ET AL.: "CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients", IMMUNITY, vol. 32, no. l, 2010, pages 129 - 40, XP009173143 |
CARTER NAVASCONCELLOS RROSSER ECTULONE CMUNOZ-SUANO AKAMANAKA M ET AL.: "Mice lacking endogenous IL-10-producing regulatory B cells develop exacerbated disease and present with an increased frequency of Thl/Thl7 but a decrease in regulatory T cells", J IMMUNOL., vol. 186, no. 10, 2011, pages 5569 - 79 |
CASARES ET AL: "A peptide inhibitor of FOXP3 impairs regulatory T cell activity and improves vaccine efficacy in mice", J IMMUNOL., vol. 185, no. 9, 1 November 2010 (2010-11-01), pages 5150 - 9, XP055075433 |
CHERUKURI AROTHSTEIN DMCLARK BCARTER CRDAVISON AHERNANDEZ-FUENTES M ET AL.: "Immunologic human renal allograft injury associates with an altered IL-10/TNF-a expression ratio in regulatory B cells", J AM SOC NEPHROL, vol. 25, no. 7, 2014, pages 1575 - 85 |
CHIORAZZI NFERRARINI M: "B cell chronic lymphocytic leukemia: lessons learned from studies of the B cell antigen receptor", ANNU REV IMMUNOL, vol. 21, 2003, pages 841 - 94, XP009096729, DOI: doi:10.1146/annurev.immunol.21.120601.141018 |
CLARK DNMARKHAM JLSLOAN CSPOOLE BD: "Cytokine inhibition as a strategy for treating systemic lupus erythematosus", CLIN IMMUNOL, vol. 148, no. 3, September 2013 (2013-09-01), pages 335 - 43, XP028688849 |
DE BENEDETTI ET AL., J IMMUNOL, vol. 166, 2001, pages 4334 - 4340 |
DEGLESNE PACHEVALLIER NLETESTU RBARAN-MARSZAK FBEITAR TSALANOUBAT C ET AL.: "Survival response to B-cell receptor ligation is restricted to progressive chronic lymphocytic leukemia cells irrespective of Zap70 expression", CANCER RES., vol. 66, no. 14, 2006, pages 7158 - 66 |
DILILLO DJWEINBERG JBYOSHIZAKI AHORIKAWA MBRYANT JMIWATA Y ET AL.: "Chronic lymphocytic leukemia and regulatory B cells share IL-10 competence and immunosuppressive function", LEUKEMIA, vol. 27, no. 1, 2013, pages 170 - 82, XP055263785 |
DRENNAN SD'AVOLA AGAO YWEIGEL CCHRYSOSTOMOU ESTEELE AJ ET AL.: "IL-10 production by CLL cells is enhanced in the anergic IGHV mutated subset and associates with reduced DNA methylation of the IL10 locus", LEUKEMIA, 2017 |
EICHHORST BROBAK TMONTSERRAT EGHIA PHILLMEN PHALLEK M ET AL.: "Chronic lymphocytic leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up", ANN ONCOL, vol. 26, no. 5, 2015, pages 78 - 84 |
EPRATUZUMAB, LEONARD ET AL., CLINICAL CANCER RESEARCH, vol. 10, pages 53Z7 - 5334 |
FABBRI GDALLA-FAVERA R: "The molecular pathogenesis of chronic lymphocytic leukaemia", NAT REV CANCER, vol. 16, no. 3, 2016, pages 145 - 62 |
FLORES-BORJA FBOSMA ANG DREDDY VEHRENSTEIN MRISENBERG DA ET AL.: "CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation", SCI TRANSL MED, vol. 5, no. 173, 2013, pages 173ra23 |
FULTON ET AL., CLINICAL CHEMISTRY, vol. 43, no. 9, 1997, pages 1749 - 1756 |
FULWYLERMCHUGH, METHODS IN CELL BIOLOGY, vol. 33, 1990, pages 613 - 629 |
GARAUD SMORVA ALEMOINE SHILLION SBORDRON APERS JO ET AL.: "CD5 promotes IL-10 production in chronic lymphocytic leukemia B cells through STAT3 and NFAT2 activation", J IMMUNOL, vol. 186, no. 8, 2011, pages 4835 - 44 |
GOROSITO SERRAN MFIOCCA VERNENGO FBECCARIA CGACOSTA RODRIGUEZ EVMONTES CLGRUPPI A: "The regulatory role of B cells in autoimmunity, infections and cancer: Perspectives beyond IL10 production", FEBS LETT, vol. 589, no. 22, 2015, pages 3362 - 9 |
GRIFFIN DOHOLODICK NEROTHSTEIN TL: "Human B1 cells in umbilical cord and adult peripheral blood express the novel phenotype CD20+ CD27+ CD43+ CD70", J EXP MED., vol. 208, no. 1, 2011, pages 67 - 80 |
HAGN MBLACKWELL SEBEYER TEBEL VFABRICIUS DLINDNER S ET AL.: "B-CLL cells acquire APC- and CTL-like phenotypic characteristics after stimulation with CpG ODN and IL-21", INT IMMUNOL, vol. 26, no. 7, 2014, pages 383 - 95 |
HALLEK MCHESON BDCATOVSKY DCALIGARIS-CAPPIO FDIGHIERO GDOHNER H ET AL.: "Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines", BLOOD, vol. 111, no. 12, 2008, pages 5446 - 56 |
HAMBLIN TJOSCIER DG: "Chronic lymphocytic leukaemia: the nature of the leukaemic cell", BLOOD REV, vol. 11, no. 3, 1997, pages 119 - 28 |
HERNDON TMCHEN SSSABA NSVALDEZ JEMSON CGATMAITAN M ET AL.: "Direct in vivo evidence for increased proliferation of CLL cells in lymph nodes compared to bone marrow and peripheral blood", LEUKEMIA, vol. 31, no. 6, 2017, pages 1340 - 7 |
HORIKAWA MMINARD-COLIN VMATSUSHITA TTEDDER TF: "Regulatory B cell production of IL-10 inhibits lymphoma depletion during CD20 immunotherapy in mice", J CLIN INVEST, vol. 121, no. 11, 2011, pages 4268 - 80 |
HOSSAIN DMPANDA AKMANNA AMOHANTY SBHATTACHARJCC PBHATTACHARYYA S ET AL.: "FoxP3 acts as a cotranscription factor with STAT3 in tumor-induced regulatory T cells", IMMUNITY, vol. 39, no. 6, 2013, pages 1057 - 69 |
INOUE SMAI ADYER MJCOHEN GM: "Inhibition of histone deacetylase class I but not class II is critical for the sensitization of leukemic cells to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis", CANCER RES, vol. 66, no. 13, 2006, pages 6785 - 92, XP002494873 |
IWATA YMATSUSHITA THORIKAWA MDILILLO DJYANABA KVENTURI GM ET AL.: "Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells", BLOOD, vol. 117, no. 2, 2011, pages 530 - 41, XP055116002 |
JAK MMOUS RREMMERSWAAL EBSPIJKER RJASPERS AYAGUE A ET AL.: "Enhanced formation and survival of CD4+ CD25hi Foxp3+ T-cells in chronic lymphocytic leukemia", LEUK LYMPHOMA, vol. 50, no. 5, 2009, pages 788 - 801 |
JITSCHIN RBRAUN MBUTTNER MDETTMER-WILDE KBRICKS JBERGER J ET AL.: "CLL-cells induce IDOhi CD14+HLA-DRlo myeloid-derived suppressor cells that inhibit T-cell responses and promote TRegs", BLOOD, vol. 124, no. 5, 2014, pages 750 - 60 |
KIPPS TJSTEVENSON FKWU CJCROCE CMPACKHAM GWIERDA WG ET AL.: "Chronic lymphocytic leukaemia", NAT REV DIS PRIMERS, vol. 3, 2017, pages 17008 |
KLEIN UTU YSTOLOVITZKY GAMATTIOLI MCATTORETTI GHUSSON H ET AL.: "Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells", J EXP MED, vol. 194, no. 11, 2001, pages 1625 - 38, XP009051394 |
KOHLERMILSTEIN, NATURE, vol. 256, 1975, pages 495 |
KRIEGLER: "A Laboratory Manual", 1990, W.H. FREEMAN C.O. |
LARSEN MARNAUD LHIE MPARIZOT CDORGHAM KSHOUKRY M ET AL.: "Multiparameter grouping delineates heterogeneous populations of human IL-17 and/or IL-22 T-cell producers that share antigen specificities with other T-cell subsets", EUR J IMMUNOL, vol. 41, no. 9, 2011, pages 2596 - 605 |
LE ROY CDEGLESNE PACHEVALLIER NBEITAR TECLACHE VQUETTIER M ET AL.: "The degree of BCR and NFAT activation predicts clinical outcomes in chronic lymphocytic leukemia", BLOOD, vol. 120, no. 2, 2012, pages 356 - 65 |
LEMOINE SMORVA AYOUINOU PJAMIN C: "Human T cells induce their own regulation through activation of B cells", J AUTOIMMUN, vol. 36, no. 3-4, 2011, pages 228 - 38, XP028226360 |
LINDNER SDAHLKE KSONTHEIMER KHAGN MKALTENMEIER CBARTH TF ET AL.: "Interleukin 21-induced granzyme B-expressing B cells infiltrate tumors and regulate T cells", CANCER RES, vol. 73, no. 8, 2013, pages 2468 - 79 |
LLORENTE ET AL.: "Clinical and biologic effects of antiinterleukin-10 monoclonal antibody administration in systemic lupus erythematosus", ARTHRITIS RHEUM, vol. 43, no. 8, 2000, pages 1790 - 1800, XP009146922 |
MARKLIN MHEITMANN JSFUCHS ARTRUCKENMULLER FMGUTKNECHT MBUGL S ET AL.: "NFAT2 is a critical regulator of the anergic phenotype in chronic lymphocytic leukaemia", NAT COMMUN, vol. 8, no. 1, 2017, pages 755 |
MATSUMOTO MBABA AYOKOTA TNISHIKAWA HOHKAWA YKAYAMA H ET AL.: "Interleukin-10-producing plasmablasts exert regulatory function in autoimmune inflammation", IMMUNITY, vol. 41, no. 6, 2014, pages 1040 - 51 |
MAURI CMENON M: "The expanding family of regulatory B cells", INT IMMUNOL, vol. 27, no. 10, 2015, pages 479 - 86, XP055485715 |
MURRY: "Methods in Molecular Biology", vol. 7, 1991, HUMANA PRESS, INC. |
MUZIO MAPOLLONIO BSCIELZO CFRENQUELLI MVANDONI IBOUSSIOTIS V ET AL.: "Constitutive activation of distinct BCR-signaling pathways in a subset of CLL patients: a molecular signature of anergy", BLOOD, vol. 112, no. 1, 2008, pages 188 - 95 |
NICHOLAS P. RESTIFOMARK E. DUDLEYSTEVEN A. ROSENBERG: "Adoptive immunotherapy for cancer: harnessing the T cell response", NATURE REVIEWS IMMUNOLOGY, vol. 12, April 2012 (2012-04-01), XP055034896 |
NOH JCHOI WSNOH GLEE JH: "Presence of Foxp3-expressing CD19(+)CD5(+) B Cells in Human Peripheral Blood Mononuclear Cells: Human CD19(+)CD5(+)Foxp3(+) Regulatory B Cell (Breg", IMMUNE NETW, vol. 10, no. 6, 2010, pages 247 - 9 |
NOUEL APOCHARD PSIMON QSEGALEN ILE MEUR YPERS JO ET AL.: "B-Cells induce regulatory T cells through TGF-p/IDO production in A CTLA-4 dependent manner", J AUTOIMMUN, vol. 59, 2015, pages 53 - 60 |
PAREKH VVPRASAD DVBANERJEE PPJOSHI BNKUMAR AMISHRA GC: "B cells activated by lipopolysaccharide, but not by anti-Ig and anti-CD40 antibody, induce anergy in CD8+ T cells: role of TGF-beta 1", J IMMUNOL., vol. 170, no. 12, 2003, pages 5897 - 911 |
PODHORECKA MDMOSZYNSKA AROLINSKI JWASIK E: "T type 1/type 2 subsets balance in B-cell chronic lymphocytic leukemia--the three-color flow cytometry analysis", LEUK RES, vol. 26, no. 7, 2002, pages 657 - 60 |
RICHES JCRAMSAY AGGRIBBEN JG: "T-cell function in chronic lymphocytic leukaemia", SEMIN CANCER BIOL, vol. 20, no. 6, 2010, pages 431 - 8, XP027545027 |
ROSENWALD AALIZADEH AAWIDHOPF GSIMON RDAVIS REYU X ET AL.: "Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia", J EXP MED., vol. 194, no. 11, 2001, pages 1639 - 47, XP009051393 |
ROSSER ECMAURI C: "Regulatory B cells: origin, phenotype, and function", IMMUNITY, vol. 42, no. 4, 2015, pages 607 - 12, XP055487943, DOI: doi:10.1016/j.immuni.2015.04.005 |
SANBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS |
SHALAPOUR SFONT-BURGADA JDI CARO GZHONG ZSANCHEZ-LOPEZ EDHAR D ET AL.: "Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy", NATURE, vol. 521, no. 7550, 2015, pages 94 - 8, XP055392755 |
SUZUKI ET AL., EUROP J OF IMMUNOL, vol. 22, no. 8, 1992, pages 1989 - 1993 |
TAYLOR JGGRIBBEN JG: "Microenvironment abnormalities and lymphomagenesis: Immunological aspects", SEMIN CANCER BIOL, vol. 34, 2015, pages 36 - 45, XP029283642 |
TERESA LOZANOMARTA ET AL: "Blockage of FOXP3 transcription factor dimerization and FOXP3/AML 1 interaction inhibits T regulatory cell activity: sequence optimization of a peptide inhibitor", ONCOTARGET, vol. 8, no. 42, 22 September 2017 (2017-09-22), pages 71709 - 71724 |
VADASZ ZPERI REIZA NSLOBODIN GBALBIR-GURMAN ATOUBI E: "The Expansion of CD25 high IL-10 high FoxP3 high B Regulatory Cells Is in Association with SLE Disease Activity", J IMMUNOL RES, 2015, pages 254245 |
WU ET AL., MOL. BIOL., vol. 294, 1999, pages 151 |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113252900A (en) * | 2021-06-19 | 2021-08-13 | 山东第一医科大学附属省立医院(山东省立医院) | ApoA-based chronic lymphocytic leukemia prognosis risk assessment system and application thereof |
CN113252900B (en) * | 2021-06-19 | 2022-04-08 | 山东第一医科大学附属省立医院(山东省立医院) | ApoA-based chronic lymphocytic leukemia prognosis risk assessment system and application thereof |
CN115261469A (en) * | 2022-06-23 | 2022-11-01 | 山东第一医科大学附属省立医院(山东省立医院) | Application of BRD9 in the diagnosis and treatment of chronic lymphocytic leukemia |
CN115261469B (en) * | 2022-06-23 | 2023-03-31 | 山东第一医科大学附属省立医院(山东省立医院) | Application of BRD9 in diagnosis and treatment of chronic lymphocytic leukemia |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20230183346A1 (en) | Antibodies having specificity for btla and uses thereof | |
US20070081989A1 (en) | Treatment of B cell diseases using anti-germline antibody binding agents | |
WO2016059220A1 (en) | Tcr-activating agents for use in the treatment of t-all | |
UA121112C2 (en) | Anti-ox40 antibodies and methods of use | |
US20230088070A1 (en) | Use of il-1beta binding antibodies | |
WO2019106126A1 (en) | Mdm2 modulators for the diagnosis and treatment of liposarcoma | |
WO2019234221A1 (en) | Methods for stratification and treatment of a patient suffering from chronic lymphocytic leukemia | |
EP3341732A1 (en) | Methods for predicting the survival time of patients suffering from a lung cancer | |
WO2019211370A1 (en) | Methods and pharmaceutical compositions for treating cancer | |
US20240262909A1 (en) | Tim-3 modulates anti-tumor immunity by regulating inflammasome activation | |
WO2018189403A1 (en) | Methods and pharmaceutical compositions for the treatment of cancer | |
EP4427044A1 (en) | Methods and compositions for treating triple negative breast cancer (tnbc) | |
EP4347043A1 (en) | Method for treating cd127-positive cancers by administering an anti-cd127 agent | |
US20210047696A1 (en) | Methods and pharmaceutical compositions for treating cancer | |
US20210355222A1 (en) | Methods of Treating Cancer | |
EP3800201A1 (en) | Cd28h stimulation enhances nk cell killing activities | |
US20220025036A1 (en) | Use of il-1beta binding antibodies | |
US20210164984A1 (en) | Methods for predicting outcome and treatment of patients suffering from prostate cancer or breast cancer | |
US20230250173A1 (en) | Biomarkers for pd-1 axis binding antagonist therapy | |
US20200088732A1 (en) | Methods for the diagnosis and treatment of pancreatic ductal adenocarcinoma | |
US20220290151A1 (en) | Use of müllerian inhibiting substance inhibitors for treating cancer | |
US20230227565A1 (en) | Method for Treating CD127-Positive Cancers by Administering an Anti-CD127 Agent | |
US20240316061A1 (en) | Bet inhibitors for treating pab1 deficient cancer | |
WO2025068393A1 (en) | Methods for the treatment of fibrotic related diseases | |
US20180201687A1 (en) | Antibodies having specificity to myosin 18a and uses thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 19728455 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 19728455 Country of ref document: EP Kind code of ref document: A1 |